US8486973B2 - Sublingual fentanyl spray - Google Patents

Sublingual fentanyl spray Download PDF

Info

Publication number
US8486973B2
US8486973B2 US12/221,333 US22133308A US8486973B2 US 8486973 B2 US8486973 B2 US 8486973B2 US 22133308 A US22133308 A US 22133308A US 8486973 B2 US8486973 B2 US 8486973B2
Authority
US
United States
Prior art keywords
fentanyl
dose
mean
formulation
mcg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US12/221,333
Other versions
US20090176834A1 (en
Inventor
S. George Kottayil
Venkat R. Goskonda
Zhongyuan Zhu
Linet Kattookaran
Neha Parikh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Btcp Pharma LLC
Original Assignee
Insys Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
US case filed in Delaware District Court litigation Critical https://portal.unifiedpatents.com/litigation/Delaware%20District%20Court/case/1%3A18-cv-01308 Source: District Court Jurisdiction: Delaware District Court "Unified Patents Litigation Data" by Unified Patents is licensed under a Creative Commons Attribution 4.0 International License.
US case filed in Delaware District Court litigation https://portal.unifiedpatents.com/litigation/Delaware%20District%20Court/case/1%3A18-cv-00414 Source: District Court Jurisdiction: Delaware District Court "Unified Patents Litigation Data" by Unified Patents is licensed under a Creative Commons Attribution 4.0 International License.
US case filed in Delaware District Court litigation https://portal.unifiedpatents.com/litigation/Delaware%20District%20Court/case/1%3A17-cv-01303 Source: District Court Jurisdiction: Delaware District Court "Unified Patents Litigation Data" by Unified Patents is licensed under a Creative Commons Attribution 4.0 International License.
First worldwide family litigation filed litigation https://patents.darts-ip.com/?family=40305148&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US8486973(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US12/221,333 priority Critical patent/US8486973B2/en
Application filed by Insys Therapeutics Inc filed Critical Insys Therapeutics Inc
Assigned to INSYS THERAPEUTICS, INC. reassignment INSYS THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GOSKONDA, VENKAT R., PARIKH, NEHA, KOTTAYIL, S. GEORGE, KATTOOKARAN, LINET, ZHU, ZHONGYUAN
Publication of US20090176834A1 publication Critical patent/US20090176834A1/en
Priority to US13/895,111 priority patent/US8835459B2/en
Publication of US8486973B2 publication Critical patent/US8486973B2/en
Application granted granted Critical
Priority to US14/448,752 priority patent/US9241935B2/en
Assigned to INSYS PHARMA, INC. reassignment INSYS PHARMA, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: INSYS THERAPEUTICS, INC.
Priority to US14/966,301 priority patent/US9642844B2/en
Assigned to INSYS DEVELOPMENT COMPANY, INC. reassignment INSYS DEVELOPMENT COMPANY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INSYS PHARMA, INC.
Priority to US15/478,499 priority patent/US10610523B2/en
Assigned to BTCP PHARMA, LLC reassignment BTCP PHARMA, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INSYS DEVELOPMENT COMPANY, INC., INSYS THERAPEUTICS, INC.
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4468Non condensed piperidines, e.g. piperocaine having a nitrogen directly attached in position 4, e.g. clebopride, fentanyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids

Definitions

  • the present invention is directed to a method of treating pain comprising sublingually administering a liquid spray formulation in the form of discrete liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns, to a human patient experiencing pain, said liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier.
  • the invention is also directed to a unit dose or bi-dose device for sublingual administration of a drug comprising:
  • the invention is further directed to a sublingual fentanyl spray formulation that further comprises water.
  • the formulations further comprise dehydrated alcohol.
  • the invention is also directed to a method of treatment wherein the sublingual formulation provides a substantially dose proportional mean AUClast based on a mean AUClast of about 4.863+/ ⁇ 1.70821 hr*ng/mL for a 400 mcg fentanyl dose when administered to humans.
  • the formulations of the present invention provide an area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration (AUC T ) of fentanyl of about 4333 pg ⁇ h/ml to about 6501 pg ⁇ h/ml based on a 800 ⁇ g fentanyl dose, preferably about 4875 pg ⁇ h/ml to about 5960 pg ⁇ h/ml based on a 800 ⁇ g fentanyl dose, more preferably about 5146 pg ⁇ h/ml to about 5689 pg ⁇ h/ml based on a 800 ⁇ g fentanyl dose after sublingual administration to humans.
  • AUC T area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration
  • the formulation is preferably included into a liquid carrier for the delivery of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, via a spray device.
  • Droplet size distribution can be determined by utilizing any reliable method known to one of skill in the art.
  • One such method uses laser diffraction devices, such as, for example, the Malvern Spraytec® with RT Sizer software.
  • a Malvern Mastersizer S by Malvern Instruments Limited (U.K.), device may also be used to determine size distribution.
  • a Malvern Mastersize S is a modular particle size analyzer offering measurement versatility. It can measure spray droplet size as well as wet and dry samples. Particles from sub-micron to a few millimeters may be measured with the Malvern Mastersizer S.
  • the mean C max value of the 100 ⁇ g dose was 172.0 pg/ml while for the 400 ⁇ g dose, it was 708.0 pg/ml and for the 800 ⁇ g dose, it was 1270.4 pg/ml.
  • the mean C max value of the 400 ⁇ g dose was 177.0 pg/ml and for the 800 pg dose, it was 158.8 pg/ml.

Abstract

The present invention is directed to sublingual formulations containing fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, suitable for administration to a patient, and methods for treatment with the formulations.

Description

This application claims the benefit of U.S. Provisional Patent Application Nos. 60/963,076, filed on Aug. 2, 2007 and 60/963,253 filed Aug. 3, 2007; the disclosures of which are hereby incorporated by reference in their entireties.
FIELD OF THE INVENTION
The invention is directed to sublingual formulations containing fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, suitable for administration to humans, and methods for treatment with the sublingual formulations.
BACKGROUND OF THE INVENTION
Fentanyl is a μ-opioid receptor agonist with analgesic potency approximately 80-100 times that of morphine. In clinical settings, fentanyl exerts its principal pharmacologic effects on the central nervous system. Its primary actions are analgesic and sedation.
The analgesic effects of fentanyl are related to the blood level of the drug. In general, the minimum effective concentration and the concentration at which toxicity occurs rise with increasing tolerance to any and all opioids. The rate of development of tolerance may vary widely among individuals. All opioid mu-receptor agonists, including fentanyl, produce dose dependent respiratory depression. The risk of respiratory depression is typically less in patients receiving chronic opioid therapy who develop tolerance to respiratory depression and other opioid effects. Serious or fatal respiratory depression can occur, even at recommended doses, in vulnerable individuals.
Orally administered fentanyl is subject to first pass effect metabolism as upwards of 50% or more of orally administered fentanyl is not absorbed. Other forms of delivery such a parenteral, buccal, and transdermal have been utilized to decrease or avoid this first pass effect for fentanyl.
Fentanyl is currently available in injectable form, as a lozenge (e.g. Actiq®), and as a transdermal system (e.g., Duragesic® 25, 50, 75, and 100 μg of fentanyl per hour). Duragesic® provides continuous systemic delivery of fentanyl for approximately 72 hours. Duragesic® is indicated in the management of chronic pain in patients requiring continuous opioid analgesia for pain that is not optimally managed with lesser means such as acetaminophen-opioid combinations, non-steroidal analgesics, or prn (as needed) dosing with short-acting opioids. Duragesic® is typically not suitable for patients experiencing acute pain due to the delay in absorption of the fentanyl through the patch, or postoperative pain because serious or life-threatening hypoventilation could result.
Actiq® is a solid formulation of fentanyl citrate, intended for oral transmucosal administration. Actiq® is a lozenge attached to a handle similar in shape to a lollipop. The handle is purportedly to allow the Actiq® unit to be removed from the mouth if signs of excessive opioid effects appear during administration. Actiq® is indicated for the management for breakthrough cancer pain in patients with malignancies who are already receiving and who are tolerant to opioid therapy for their underlying persistent cancer pain. Actiq® is contraindicated in the management of acute or postoperative pain.
Sublingual tablets and lozenges (e.g., Actiq®) which may be used for acute pain or breakthrough pain have certain disadvantages. A disadvantage, amongst others, is that after intake the active agent in these pharmaceutical agents must first be released and dispersed prior to being available for resorption in dissolved form. In addition, the absorption pharmacokinetics of fentanyl from Actiq® may vary depending on the fraction of the dose that is absorbed through the oral mucosa and the fraction swallowed. Further, certain lozenges may be in the form of a candy which require medical supervision and may be socially questionable.
There exists a need in art for a sublingual formulation including fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, which is suitable for effective pain management.
SUMMARY AND OBJECTS OF THE INVENTION
It is an object of the invention to provide a fentanyl formulation suitable for sublingual administration for effective pain management.
It is an object of certain embodiments of the invention to provide methods and compositions capable of rapidly inducing a state of sedation, analgesia, and/or anesthesia.
It is a further object of certain embodiments of the invention to provide methods and compositions for fentanyl administration which minimize the underdosing and/or overdosing of a patient in need of fentanyl therapy.
It is a further object of certain embodiments of the invention to provide methods and compositions suitable for the treatment of breakthrough pain in patients receiving chronic pain treatment.
It is a further object of certain embodiments of the present invention to provide a method for sublingual administration of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, in a controlled amount for the treatment of pain.
It is a further object of certain embodiments of the present invention to provide a dosage form of an opioid analgesic which can be administered sublingually in a manner which will cause substantial sublingual absorption without substantial risk of the dose passing into the lungs of the recipient.
The above-mentioned objects and others are achieved by virtue of the present invention, which is directed in part to a method for sublingually administering fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, to provide fast-acting relief in a formulation in which a substantial portion of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof will not be passed into the lungs of the patient.
In certain embodiments the present invention is directed to a sublingual fentanyl formulation comprising discrete liquid droplets comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, said droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns.
In certain embodiments, the present invention is directed to a sublingual fentanyl formulation comprising discrete liquid droplets of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; in a pharmaceutically acceptable liquid carrier; said droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
In certain preferred embodiments, none of the particles have a diameter which would allow the fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof to be delivered to the lung upon sublingual administration.
In certain embodiments, the present invention is directed to a unit dose of a sublingual fentanyl formulation, said unit dose comprising discrete liquid droplets of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; and a pharmaceutically acceptable liquid carrier; said droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns.
In certain embodiments, the present invention is directed to a unit dose of a sublingual fentanyl formulation, said unit dose comprising discrete liquid droplets of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; and a pharmaceutically acceptable liquid carrier; said droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
In certain embodiments, the present invention is directed to a method of treating pain comprising sublingually administering a liquid spray formulation in the form of discrete liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns, to a human patient experiencing pain, said liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier.
In certain embodiments, the present invention is directed to a method of treating pain comprising sublingually administering a liquid spray formulation in the form of discrete liquid droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns to a human patient experiencing pain; said liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier.
In certain embodiments, the present invention is directed to a device which includes a reservoir containing a unit dose of a liquid formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof in a pharmaceutically acceptable liquid carrier; the device having an actuator which when actuated delivers the unit dose of the liquid formulation in the form of liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns. Preferably, the device delivers a therapeutically effective dose of the liquid formulation in the form of liquid droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
In certain embodiments, the present invention is directed to a multi-dose device which includes a reservoir containing a liquid formulation comprising fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof in a pharmaceutically acceptable liquid carrier; the device having an actuator which when actuated delivers a therapeutically effective dose of the liquid formulation in the form of liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns. Preferably, the device delivers a therapeutically effective dose of the liquid formulation in the form of liquid droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
In certain embodiments, the present invention is directed to a method of treating pain comprising utilizing a spray device which includes a reservoir including a liquid formulation comprising fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof in a pharmaceutically acceptable liquid carrier; and an actuator which upon actuation delivers a therapeutically effective amount of liquid droplets to be sprayed from the device having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns.
In certain embodiments, the present invention is directed to a method of treating pain comprising utilizing a spray device which includes a reservoir including a liquid formulation comprising fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; and a pharmaceutically acceptable liquid carrier; and an actuator which upon actuation delivers a therapeutically effective amount of liquid droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
In certain embodiments, the present invention is directed to a method of treating breakthrough pain comprising sublingually administering a liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier to a human patient to treat breakthrough pain experienced by said human patient.
In certain embodiments, the present invention is directed to a method of treating breakthrough pain comprising sublingually administering a liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier to a human patient who is receiving chronic pain treatment, and is experiencing breakthrough pain.
In certain embodiments, the present invention is directed to a method of reducing patient to patient variability for the treatment of breakthrough pain, comprising sublingually administering to a human patient experiencing breakthrough pain a dose of fentanyl in a liquid spray formulation comprising fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, and a pharmaceutically acceptable liquid carrier said liquid spray formulation being administered as discrete liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns.
In certain embodiments, the present invention is directed to a method of reducing patient to patient variability for the treatment of breakthrough pain, comprising sublingually administering to a human patient experiencing breakthrough pain a dose of fentanyl in a liquid spray formulation comprising fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, and a pharmaceutically acceptable liquid carrier said liquid spray formulation being administered as discrete liquid droplets having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns.
In certain preferred embodiments, the liquid spray formulation further includes a pharmaceutically acceptable solvent. Preferably the pharmaceutically acceptable solvent is an organic solvent which is included in an amount suitable for dissolving the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof.
In certain preferred embodiments the formulations of the present invention provide a mean time to maximum plasma concentration (Tmax) of fentanyl at from about 5 minutes to about 120 minutes, after sublingual administration to humans.
In certain preferred embodiments the formulations of the present invention provide a mean maximum plasma concentration (Cmax) of fentanyl of about 127 pg/ml to about 213 pg/ml per 100 μg fentanyl after sublingual administration to humans.
In certain preferred embodiments of the present invention the formulations of the present invention do not include a propellant.
In certain embodiments, the formulations of the present invention are suitable for transmucosal administration, including, for example, buccal administration.
In certain embodiments, the present invention is further directed to a method of transmucosally administering fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, to a human to provide fast-acting relief in a formulation in which a substantial portion of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof will not be passed into the lungs of the patient. In certain preferred embodiments, the transmucosal area is the buccal area of a human.
In certain embodiments, the present invention is further directed to the use of a formulation as defined above for the manufacture of a medicament for use as an analgesic, for the treatment of acute pain and/or breakthrough pain, as an anesthetic premedication, for the induction of anesthesia, as a sedative and/or for the treatment of anxiety.
The invention is also directed to a sublingual fentanyl formulation comprising discrete liquid droplets of an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, in a pharmaceutically acceptable liquid carrier; said droplets having a mean diameter of at least about 10 microns, and upon administration to a human patient, at least about 90% of the discrete liquid droplets have a mean diameter equal or greater than about 9 μm. In other embodiments, not more than about 5% of the discrete liquid droplets have a mean diameter less than 9 μm. In still other embodiments, the formulation provides a respirable dose of not more than about 5% of the total fentanyl dose contained.
The invention is also directed to a method of treating pain comprising sublingually administering a liquid spray formulation in the form of discrete liquid droplets having a mean diameter of at least about 10 microns to a human patient experiencing pain and at least about 90% of the discrete liquid droplets have a mean diameter equal or greater than about 9 μm upon administration to a human patient, said liquid spray formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, dispersed in a pharmaceutically acceptable liquid carrier. In certain other embodiments, not more than about 5% of the discrete liquid droplets have a mean diameter less than 9 μm. In other embodiments, the formulation provides a respirable dose of not more than about 5% of the total fentanyl dose contained.
The invention is also directed to a unit dose or bi-dose device for sublingual administration of a drug comprising:
a reservoir containing a unit dose or a bi-dose of a liquid formulation comprising an effective amount of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof in a pharmaceutically acceptable liquid carrier; and
the device having an actuator which when actuated delivers the unit dose of the liquid formulation in the form of liquid droplets having a mean diameter of at least about 10 microns, and wherein upon administration to a human patient, at least about 90% of the discrete liquid droplets have a mean diameter equal or greater than about 9 μm.
In other embodiments, not more than about 5% of the discrete liquid droplets have a mean diameter less than 9 μm. In still other embodiments, the formulation provides a respirable dose of not more than about 5% of the total fentanyl dose contained.
In accordance with the above objects, the invention is also directed to a sublingual spray formulation comprising an effective amount of fentanyl and at least one pharmaceutically acceptable excipient, the formulation providing a mean Tmax of about 1.28+/−0.60 hours when a dose is administered sublingually to humans. In certain other embodiments, the sublingual formulation has a concentration of fentanyl from about 1 mg/mL to about 8 mg/mL. In certain preferred embodiments, the concentration of fentanyl is about 1 mg/mL, about 2 mg/mL, about 4 mg/mL, about 6 mg/mL or about 8 mg/mL.
In accordance with certain of the above objections, the invention is also directed to a sublingual formulation exhibiting a mean Cmax of about 0.813 ng/ml+/−0.252 based on a sublingual dose of about 400 mcg fentanyl when administered to humans.
In certain other embodiments, the sublingual formulation provides a dosage amount of fentanyl selected from the group consisting of about 100 mcg, about 200 mcg, about 600 mcg and about 800 mcg, and provides a mean Cmax which is substantially dose proportional to the sublingual formulation containing 400 mcg fentanyl dosage amount, when administered to humans.
In still other embodiments, the sublingual formulation provides a substantially dose proportional mean Cmax based on a mean Cmax of about 0.813 ng/ml+/−0.252 for a 400 mcg fentanyl dose when administered to humans.
The present invention also provides a sublingual formulation which provides a mean Tmax when administered to humans selected from the group consisting of: about 1.12 hours when the formulation provides a 100 mcg dose, about 1.04 hours when the formulation provides a 200 mcg dose, about 0.97 hours when the formulation provides a 400 mcg dose, about 0.987 hours when the formulation provides a 600 mcg dose, and about 1.06 hours when the formulation provides a 800 mcg dose.
In accordance with the above objects, it is a further object of the invention to provide a sublingual fentanyl formulation which provides a plasma concentration after administration to humans selected from the group consisting of: about 60% of the mean Cmax in about 10 minutes, about 86% of the mean Cmax by about 20 minutes and a combination thereof.
In other embodiments, the invention is directed to a sublingual fentanyl formulation that when administered to humans provides a plasma concentration that is greater than about 80% of the mean Cmax for about 2 hours.
In still other embodiments, the sublingual formulation comprises 400 mcg of fentanyl, providing one or more mean pharmacokinetic values selected from the group consisting of: AUClast 4.863+/−1.70821 hr*ng/mL, AUCinf 5.761+/−1.916 hr*ng/mL, and AUCextrap 10.26+/−5.66%, when administered to humans.
In even still further embodiments of the invention, the sublingual fentanyl formulation provides a dosage amount of fentanyl when administered to humans which is substantially dose proportional to the dosage which contains about 400 mcg fentanyl selected from the group consisting of about 100 mcg, about 200 mcg, about 600 mcg, about 800 mcg, and provides one or more pharmacokinetic values selected from the group consisting of: mean AUClast, mean AUCinf, and mean AUCextrap.
In still further embodiments of the invention, the sublingual fentanyl formulation provides a substantially dose proportional mean AUClast based on a mean AUClast of about 4.863+/−1.70821 hr*ng/mL for a 400 mcg fentanyl dose when administered to humans.
It is also an object of the invention to a provide a sublingual formulation comprising a 400 mcg dose of fentanyl, providing a geometric mean ln(Cmax) of about 0.7865 ng/ml when a dose is administered to humans.
In still other embodiments, the sublingual formulation comprises a 400 mcg dose of fentanyl, providing a mean F(AUClast) of about 0.721+/−0.199 ng/mL when a dose is administered to humans.
In other embodiments, the invention is directed to a sublingual formulation comprising a 400 mcg dose of fentanyl that when administered to humans, provides a mean F (bioavailability) selected from the group consisting of: about 71%+/−16%, 0.721+/−0.199 based on AUClast and about 0.756+/−0.212 based on AUCinf, or combinations thereof.
In accordance with the above objects, the invention is further directed to a sublingual fentanyl spray formulation that further comprises water. In other embodiments, the formulations further comprise dehydrated alcohol.
It is also an object of the invention to provide a method of treating pain comprising administering to a patient in need thereof a sublingual spray formulation comprising an effective amount of fentanyl and at least one pharmaceutically acceptable excipient, the formulation providing a mean Tmax of about 1.28+/−0.60 hours when a dose is administered sublingually to humans.
In accordance with the above objects, the invention is also directed to a method of treatment wherein the sublingual formulation provides a substantially dose proportional mean Cmax based on a mean Cmax of about 0.813 ng/ml+/−0.252 for a 400 mcg fentanyl dose when administered to humans.
In yet further embodiments, the invention is also directed to a method of treatment wherein the sublingual formulation provides a substantially dose proportional mean AUClast based on a mean AUClast of about 4.863+/−1.70821 hr*ng/mL for a 400 mcg fentanyl dose when administered to humans.
It is a further object of the invention to provide a method of manufacturing a sublingual spray formulation comprising an effective amount of fentanyl and at least one pharmaceutically acceptable excipient comprising: admixing fentanyl, purified water and dehydrated alcohol and placing the mixture into a sublingual delivery device; wherein the formulation provides a mean Tmax of about 1.28+/−0.60 hours when a dose is administered sublingually to humans and upon delivery provides particles having a mean aerodynamic particle size of at least about 10 microns.
Many patients with e.g., cancer, typically continue to experience moderate to severe pain despite chronic analgesic therapy and this can occur as intermittent breakthrough pain, often due to increases in a patient's activity level. Attempts to counteract this type of pain by increasing the dose of long-acting formulations of analgesics often produce slow onset of analgesia and unwanted side-effects of sedation, constipation or nausea and vomiting. However, in certain embodiments the present invention is directed to a formulation which preferably provides a rapidly acting, potent analgesic which reduces the pain, such as breakthrough pain, for the required time and then preferably wears off fairly quickly thereby minimizing the side-effects of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof.
For purposes of the present invention, derivatives of fentanyl include sufentanil, carfentanil, lofentanil, alfentanil, or the like.
For purposes of the present invention, “breakthrough pain” refers to a pain that exceeds a threshold in a patient which causes cognizable discomfort wherein the pain experienced by the patient is otherwise typically controlled e.g., by chronic analgesic therapy, and tolerated. For example, pain related to medical illnesses, such as cancer, typically fluctuates, and patients often report the experience of cognizable discomfort (e.g., breakthrough pain). Typically breakthrough pain is currently treated with immediate release oral dosage forms which may take up to about 45 minutes or longer for the drug to be absorbed and may result in a delay of the relief of breakthrough pain, as opposed to a liquid spray formulation of the present invention which begins to provide relief of the breakthrough pain almost immediately after administration.
For purposes of the present invention, “chronic pain treatment” refers to a daily or round-the-clock pain treatment. Chronic pain treatment can be oral, parenteral, transdermal, or other suitable means of administration.
For purposes of the present invention, “sublingual” is defined herein as beneath or concerning the area under the tongue.
For purposes of the present invention the term “sublingual administration” is defined herein as the therapeutic administration of a pharmaceutical composition under the tongue.
For purposes of the present invention an “effective amount” of a drug is an amount effective to demonstrate a desired activity of the drug. According to the instant invention, a therapeutically effective amount of fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof, is an amount effective to treat, e.g., noticeably reduce, pain in a patient.
For purposes of the present invention the terms droplets and particles may be used interchangeably.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 depicts the mean (±S.E.) plasma concentration-time profiles following intravenous administration of Fentanyl of Example 1 (n=3) in the study of Example 6.
FIG. 2 depicts the mean (±S.E.) plasma concentration-time profiles following sublingual administration of Fentanyl of Example 1 (n=3) in the study of Example 6.
FIG. 3 depicts the mean (±S.E.) plasma concentration-time profiles following intravenous administration of Fentanyl of Example 2 (n=3) in the study of Example 6.
FIG. 4 depicts the mean (±S.E.) plasma concentration-time profiles following sublingual administration of Fentanyl of Example 2 (n=3) in the study of Example 6.
FIG. 5 depicts the mean (±S.E.) plasma concentration-time profiles following intravenous administration of Fentanyl of Example 3 (n=3) in the study of Example 6.
FIG. 6 depicts the mean (±S.E.) plasma concentration-time profiles following sublingual administration of Fentanyl of Example 3 (n=3) in the study of Example 6.
FIG. 7 depicts the mean (±S.E.) plasma concentration-time profiles following intravenous administration of Fentanyl of Example 4 (n=3) in the study of Example 6.
FIG. 8 depicts the mean (±S.E.) plasma concentration-time profiles following sublingual administration of Fentanyl of Example 4 (n=3) in the study of Example 6.
FIG. 9 depicts the mean (±S.E.) plasma concentration-time profiles following intravenous administration of Fentanyl of Example 5 (n=3) in the study of Example 6.
FIG. 10 depicts the mean (±S.E.) plasma concentration-time profiles following sublingual administration of Fentanyl of Example 5 (n=3) in the study of Example 6.
FIG. 11 depicts a graphical summary of the Dv10, Dv50, Dv90, and plume records values versus placement distance for vignetting results.
FIG. 12 depicts a graphical summary of Dv10, Dv50, and Dv90 values versus placement at 4 cm device to laser beam for exhaust results.
FIG. 13 depicts a graphical summary of Dv10, Dv50, and Dv90 values versus placement at 7 cm device to laser beam for exhaust results.
FIG. 14 depicts a graphical summary of device to laser beam placement results for Dv10, Dv50, and Dv90 values versus distance to device to laser beam.
FIG. 15 depicts the mean fentanyl concentration-time profiles after administration of Fentanyl Sublingual Spray 400 mcg (Treatment A), Actiq® 400 mcg (Treatment B) and Fentanyl Citrate Injection 100 mcg (Treatment C) in the clinical study of Example 19.
FIG. 16 depicts the preliminary pharmacokinetic data in the dose escalation study of Example 20.
FIG. 17 depicts the preliminary pharmacokinetic data in the dose escalation study of Example 20.
FIG. 18 depicts the preliminary pharmacokinetic data in the dose escalation study of Example 20.
DETAILED DESCRIPTION OF THE INVENTION
The simplest and most prevalent administration route for pharmacologic agents is by mouth. To use this method, a pharmacological agent is typically incorporated into a tablet, a capsule, or into a liquid base. Oral administration of a drug is extremely convenient, and for many drugs, it will continue to be the method of choice.
Absorption of a drug into the bloodstream after swallowing a tablet may vary from patient to patient. The absorption of the drug is typically dependent upon the movement from the stomach to the small and large intestines and the effects of secretions from these organs. Further, with the oral administration of a drug such as fentanyl to a patient, as the fentanyl enters the patient's bloodstream through the intestines and passes through the patient's liver before distribution throughout the body, upwards of fifty percent or more of the fentanyl may be removed from the patient's bloodstream. This “first pass effect” results in the oral route of administration being impractical for fentanyl.
Absorption of fentanyl or a pharmaceutically acceptable salt thereof into the bloodstream following oral administration is significantly reduced by the first pass effect. Therefore, the oral route of administration is impractical for fentanyl. Other forms of delivery such a parenteral, buccal, and transdermal delivery have been utilized to decrease or avoid this first pass effect for fentanyl. However, these other forms of delivery have certain disadvantages associated with them. For example, parenteral administration requires injection using a syringe and needle, and may lead to necrosis that can accompany i.m. administration of drugs; Actiq® a transmucosal fentanyl citrate lozenge formulation requires the patient to constantly suck on the lozenge which is attached to a handle (similar to a lollipop) in order to obtain effective pain relief; and Duragesic®, a transdermal fentanyl delivery device, is suitable for the management of chronic pain, but is not indicated for acute or breakthrough pain.
The oral cavity offers a simple, painless method of opioid analgesic administration. Within the oral cavity, there are three generally recognized routes of administration of an active agent, namely local, buccal and sublingual.
Local delivery is mainly limited to applications regarding disruptions occurring within the oral cavity itself, such as a canker sore.
The buccal mucosa area encompasses the mucosal membranes of the inner lining of the cheeks. The buccal mucosa is however, less permeable than the sublingual area. One of the major disadvantages associated with buccal mucosa delivery of an active agent has been the relatively low passage of active agents across the mucosal epithelium, thereby resulting in low agent bioavailability, which translates into a substantial loss of usable active agent within each dosage.
Sublingual delivery is achieved through the mucosal membranes lining the floor of the mouth. Because of the high permeability and the rich blood supply, transport via the sublingual route results in a rapid onset of action, providing a delivery route appropriate for highly permeable drugs with short delivery period requirements and an infrequent dosing regimen.
The sublingual formulations of the present invention are useful in the treatment of moderate to severe pain. Preferably the sublingual formulations of the present invention are useful for the treatment of breakthrough pain. For example, the formulations of the present invention are preferably suitable for a patient receiving chronic pain therapy who experiences breakthrough pain and is in need of acute pain relief.
The sublingual formulations of the present invention may be used to alleviate pain from many causes, including but not limited to shock, limb amputation, severe chemical or thermal burn injury, sprains, ligament tears, fractures, wounds and other tissue injuries, dental surgery, procedures and maladies, labor and delivery, during physical therapy, post operative pain, radiation poisoning, cancer, acquired immunodeficiency syndrome (AIDS), epidural (or peridural) fibrosis, back surgery and laminectomy, sciatica, painful sickle cell crisis, arthritis, autoimmune disease, intractable bladder pain, and the like. Sublingual administration of the formulations of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, of the present invention is also preferably amenable to hospice use, particularly hospices that specialize in the care of cancer and AIDS patients.
In certain preferred embodiments, the sublingual administration of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, can relieve or alleviate episodes of acute breakthrough pain that can occur in a chronic pain condition. In a further embodiment, sublingual administration of fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof can be used as an adjunct therapy to a conventional treatment regimen for a chronic pain condition to alleviate breakthrough pain. In certain embodiments, the invention also provides a formulation for use as an anesthetic premedication, for the induction of anesthesia, for use as a sedative and/or for the treatment of anxiety.
Sublingual administration of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, in accordance with the present invention may be particularly beneficial in the patient with cancer who is unable to tolerate oral administration because of nausea and vomiting, dysphagia as a result of disease, or parenteral administration because of decreased venous access, emaciation, or coagulation defects. Sublingual administration of fentanyl in accordance with the present invention preferably has potential advantages of even greater ease of use and rapid onset of pain relief action. Furthermore, because sublingual venous drainage is systemic rather than portal, hepatic first-pass elimination may be avoided. The present invention preferably provides therapeutic formulations and methods for solutions of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof to be delivered by sublingual spray pumps.
In certain preferred embodiments, the sublingual administration of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, is advantageous over other forms of administration in that it does not require injection using a syringe and needle, it avoids necrosis that can accompany i.m. administration of drugs, and it avoids the need to constantly suck on a lozenge or lollipop. Preferably the sublingual administration of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, in accordance with the present invention is suitable for self administration.
In preferred embodiments certain embodiments, the formulations of the present invention are advantageous in that propellants such as hydrofluorocarbon propellants such as volatile chlorofluocarbons (e.g. propellant 12), volatile hydrofluoroalkanes (e.g. 1,1,1,2-tetrafluoroethane and 1,1,1,2,3,3,3-heptafluoro-n-propane) and volatile alkanes (e.g. propane, butane) are not required to deliver the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, sublingually to the patient.
Preferably the formulations of the present invention are delivered as liquid droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns. Most preferably the formulations are delivered as liquid droplets have a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
Preferably the delivery of the formulation of the present invention to the sublingual mucosa via spray results in a rapid onset of therapeutic effect of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof. For example, in certain embodiments the formulations of the present invention provide a mean time to maximum plasma concentration (Tmax) of fentanyl at from about 5 minutes to about 120 minutes, preferably at from about 10 to about 60 minutes, and more preferably at from about 15 to about 35 minutes after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide a mean maximum plasma concentration (Cmax) of fentanyl of about 127 pg/ml to about 213 pg/ml per 100 μg fentanyl, preferably about 142 pg/ml to about 195 pg/ml per 100 μg fentanyl, more preferably about 158 pg/ml to about 177 pg/ml per 100 μg fentanyl after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide a mean maximum plasma concentration (Cmax) of fentanyl of about 137 pg/ml to about 207 pg/ml based on a 100 μg fentanyl dose, preferably about 154 pg/ml to about 190 pg/ml based on a 100 μg fentanyl dose, more preferably about 163 pg/ml to about 181 pg/ml based on a 100 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide a mean maximum plasma concentration (Cmax) of fentanyl of about 566 pg/ml to about 850 pg/ml based on a 400 μg fentanyl dose, preferably about 637 pg/ml to about 779 pg/ml based on a 400 μg fentanyl dose, more preferably about 672 pg/ml to about 744 pg/ml based on a 400 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide a mean maximum plasma concentration (Cmax) of fentanyl of about 1016 pg/ml to about 1525 pg/ml based on a 800 μg fentanyl dose, preferably about 1143 pg/ml to about 1398 pg/ml based on a 800 μg fentanyl dose, more preferably about 1206 pg/ml to about 1334 pg/ml based on a 800 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve to infinity (AUC) of fentanyl of about 572 pg·h/ml to about 1273 pg·h/ml per 100 μg fentanyl, preferably about 644 pg·h/ml to about 1167 pg·h/ml per 100 μg fentanyl, more preferably about 715 pg·h/ml to about 1061 pg·h/ml per 100 μg fentanyl after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve to infinity (AUC) of fentanyl of about 654 pg·h/ml to about 982 pg·h/ml based on a 100 μg fentanyl dose, preferably about 736 pg·h/ml to about 900 pg·h/ml based on a 100 μg fentanyl dose, more preferably about 777 pg·h/ml to about 859 pg·h/ml based on a 100 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve to infinity (AUC) of fentanyl of about 3394 pg·h/ml to about 5092 pg·h/ml based on a 400 μg fentanyl dose, preferably about 3818 pg·h/ml to about 4667 pg·h/ml based on a 400 μg fentanyl dose, more preferably about 4030 pg·h/ml to about 4455 pg·h/ml based on a 400 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve to infinity (AUC) of fentanyl of about 4581 pg·h/ml to about 6873 pg·h/ml based on a 800 μg fentanyl dose, preferably about 5154 pg·h/ml to about 6300 pg·h/ml based on a 800 μg fentanyl dose, more preferably about 5440 pg·h/ml to about 6014 pg·h/ml based on a 800 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration (AUCT) of fentanyl of about 378 pg·h/ml to about 1067 pg·h/ml per 100 μg fentanyl, preferably about 425 pg·h/ml to about 978 pg·h/ml per 100 μg fentanyl, more preferably about 472 pg·h/ml to about 889 pg·h/ml per 100 pg fentanyl after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration (AUCT) of fentanyl of about 378 pg·h/ml to about 568 pg·h/ml based on a 100 μg fentanyl dose, preferably about 425 pg·h/ml to about 520 pg·h/ml based on a 100 μg fentanyl dose, more preferably about 448 pg·h/ml to about 497 pg·h/ml based on a 100 μg fentanyl dose after sublingual administration to humans.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration (AUCT) of fentanyl of about 2844 pg·h/ml to about 4268 pg·h/ml based on a 400 μg fentanyl dose, preferably about 3200 pg·h/ml to about 3912 pg·h/ml based on a 400 μg fentanyl dose, more preferably about 3378 pg·h/ml to about 3734 pg·h/ml based on a 400 μg fentanyl dose after sublingual administration to humans.
In certain embodiments, an increase in pH of the formulations increases absorption.
In certain other embodiments, an increase in temperature increases absorption.
In certain further embodiments the formulations of the present invention provide an area under the plasma concentration time curve from zero to the time of the last quantifiable plasma concentration (AUCT) of fentanyl of about 4333 pg·h/ml to about 6501 pg·h/ml based on a 800 μg fentanyl dose, preferably about 4875 pg·h/ml to about 5960 pg·h/ml based on a 800 μg fentanyl dose, more preferably about 5146 pg·h/ml to about 5689 pg·h/ml based on a 800 μg fentanyl dose after sublingual administration to humans.
Preferably the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, is dissolved in an organic solvent. Examples of organic solvents that may be used to enhance the solubility of fentanyl, or the pharmaceutically acceptable salt thereof in a carrier such as e.g., water, include for example and without limitation: lower alcohols (e.g. C1-4 alcohols) such as methanol, ethanol, propyl alcohol, or butyl alcohol; C2-8 alcohols having two or three hydroxyl groups, preferably glycerol, propylene glycol or butylene glycol; and polyethylene glycols such as PEG200 and PEG400 and the like. Mixtures of any of the aforementioned solvents may be used. In certain embodiments, the solvent is a non-polar hydrocarbon, preferably a C7-18 hydrocarbon of a linear or branched configuration, its alcohols, fatty acid esters, and triglycerides, such as miglyol. In certain preferred embodiments the organic solvent is ethanol, propylene glycol, polyethylene glycol, or combination thereof.
Preferably the amount of organic solvent for inclusion in the formulation is at least an amount of organic solvent necessary to adequately solubilize the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, such that the fentanyl remains in solution and does not precipitate out.
In certain embodiments, the organic solvent is included in the formulation in an amount of from about 0% to about 99.9% by weight of the formulation, preferably from about 10% to about 80% by weight of the formulation, more preferably from about 20% to about 60% by weight of the formulation.
In certain embodiments, the compositions comprise a C2-8 alcohol such as propylene glycol, or a polyethylene glycol and/or polypropylene glycol of an average molar weight of 200 to 4000, or a mixture thereof, in addition to the organic solvent described above. The C2-8 alcohol may act as a cosolvent in combination with the organic solvent. Polyethylene glycols commercially available as Carbowax® (e.g., Carbowax 300 of a molar weight of 300), can be used.
In certain embodiments, the solvent is a cosolvent which includes any of the solvents mentioned herein. In certain preferred embodiments, the cosolvent includes ethanol, propylene glycol, polyethylene glycol, labrosol, labrafil, transcutol, or combination thereof
In certain preferred embodiments, the composition according to the invention comprises from about 0.0001% to about 20% by weight of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; from about 1% to about 99% by weight of organic solvent; and from about 0.01% to about 50% by weight of C2-8 alcohol.
In certain preferred embodiments, the composition according to the invention comprises from about 0.001% to about 15% by weight of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; from about 5% to 90% by weight of ethanol; and from about 0.1% to 40% by weight of propylene glycol.
In certain preferred embodiments, the composition according to the invention comprises from about 0.01% to about 10% by weight of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; from about 10% to about 80% by weight of ethanol; and from about 1% to about 30% by weight of propylene glycol.
In certain preferred embodiments, the composition according to the invention comprises from about 0.1% to about 0.8% by weight of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof; from about 20% to about 60% by weight of ethanol; and from about 4% to about 6% by weight of propylene glycol.
In certain preferred embodiments, the composition according to the invention comprises in a 1 ml volume: from about 100 μg/ml to about 800 μg/ml fentanyl base, about 50% ethanol, about 5.2% propylene glycol, and water qs to 1 ml.
In certain embodiments the fentanyl is employed in the form of a pharmaceutically acceptable salt. Examples of suitable salt forms of fentanyl for use in accordance with the present invention include for example and without limitation, the hydrochloride, chloride, sulphate, tartrate, or citrate salt forms. In certain preferred embodiments, the fentanyl is employed as the free base in the formulations of the present invention.
In certain preferred embodiments, the fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof, will be employed in the formulation at a concentration of from about 0.05 mg/ml to about 15 mg/ml, preferably from about 0.1 mg/ml to about 10 mg/ml, more preferably from about 1 mg/ml to about 8 mg/ml (where weight is expressed as weight of fentanyl free base).
In certain preferred embodiments, the amount of fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof; delivered per unit dose is about 10 μg to about 10 mg, preferably from about 25 μg to about 5 mg, more preferably from about 50 μg to about 1600 μg.
In preferred embodiments of the present invention, the formulation is a solution. In certain alternate embodiments, the formulation is a suspension. When the formulation of the present invention is a suspension, it may be necessary to shake the composition prior to spraying.
In certain preferred embodiments, after the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, is dissolved in the organic solvent, the formulation is preferably included into a liquid carrier for the delivery of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, via a spray device.
Pharmaceutically acceptable carriers include but are not limited to water, buffer, saline, buffered saline, dextrose solution, propylene glycol, polyethylene glycols, miglyol, and the like. In a specific embodiment, a carrier that may be used in the pharmaceutical formulation of the present invention is phosphate buffered saline, or a buffered saline. In certain preferred embodiments the carrier is water. In certain embodiments, the water in the formulation is present in the form of an aqueous buffer. The buffer is preferably adapted to stabilize the pH of the formulation at pH of about 5 to about 12, preferably at pH of about 6 to about 10, more preferably from about 8 to about 9.5. Buffer systems for use in accordance with the present invention include for example and without limitation sodium acetate/acetic acid, ammonium acetate/disodium edentate, boric acid/sodium hydroxide, orthophosphoric acid/sodium hydroxide, sodium hydrogen carbonate/sodium carbonate, disodium hydrogen orthophosphate/citric acid, and the like.
Other components such as preservatives, antioxidants, surfactants, absorption enhancers, viscosity enhancers or film forming polymers, bulking agents, diluents, coloring agents, flavoring agents, pH modifiers, sweeteners or taste-masking agents may also be incorporated into the composition. Suitable coloring agents include red, black and yellow iron oxides and FD&C dyes such as FD&C Blue No. 2, FD&C Red No. 40, and the like. Suitable flavoring agents include mint, raspberry, licorice, orange, lemon, grapefruit, caramel, vanilla, cherry grape flavors, combinations thereof, and the like. Suitable pH modifiers include citric acid, tartaric acid, phosphoric acid, hydrochloric acid, maleic acid, sodium hydroxide, and the like. Suitable sweeteners include aspartame, acesulfame K, thaumatic, and the like. Suitable taste-masking agents include sodium bicarbonate, ion-exchange resins, cyclodextrin inclusion compounds, adsorbates, and the like.
Absorption enhancers for use in accordance with the present invention include, for example, polysorbates, sorbitan esters, poloxamer block copolymers, PEG-35 castor oil, PEG-40 hydrogenated castor oil, caprylocaproyl macrogol-8 glycerides, PEG-8 caprylic/capric glycerides, sodium lauryl sulfate, dioctyl sulfosuccinate, polyethylene lauryl ether, ethoxydiglycol, propylene glycol mono-di-caprylate, glycerol monocaprylate, glyceryl fatty acids (C8-C18) ethoxylated, oleic acid, linoleic acid, glyceryl caprylate/caprate, glyceryl monooleate, glyceryl monolaurate, caprylic/capric triglycerides, ethoxylated nonylphenols, PEG-(8-50) stearates, olive oil PEG-6 esters, triolein PEG-6 esters, lecithin, d-alpha tocopheryl polyethylene glycol 1000 succinate, polycarbonate, sodium glycocholate, sodium taurocholate, cyclodextrins, citric acid, sodium citrate, triacetin, combinations thereof, and the like. In certain preferred embodiments, the absorption enhancer is triacetin. In certain preferred embodiments wherein an absorption enhancer is included in the formulation, the absorption enhancer is included in an amount of from about 0.001% to about 10% by weight of the formulation, preferably in an amount of about 0.01% to about 5% by weight of the formulation.
Bulking agents for use in accordance with the present invention include for example, microcrystalline cellulose, mannitol, xylitol, starches and the like. In certain preferred embodiments, the bulking agent is mannitol. In certain preferred embodiments wherein bulking agent is included in the formulation, the bulking agent is included in an amount of from about 0.001% to about 10% by weight of the formulation, preferably in an amount of about 0.01% to about 5% by weight of the formulation.
Film-forming polymers for use in accordance with the present invention may serve for decreasing the fineness of the spray, the spraying angle and preferably the spreading by increasing the viscosity of the composition. As film-forming polymer, gellan gum, xantham gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, gelucire, poloxamers, alginic acid, propyleneglycol ester, polyvinyl alcohol (PVA), polyvinylpyrrolidone (PVP), PVP/PVA copolymer, lubrajel, carboxyvinyl polymer, acrylic acid polymers and copolymers, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate, ethyl methacrylate, combinations thereof and the like can be used. In certain embodiments, an increase in the viscosity of the solution using film-forming polymers or the like provides an increase in the droplet size when administered from the spray device. The chemistry of the polymer and the molecular weight of the polymer may also influence the diameter of the droplets.
In certain embodiments, the formulations according to the invention are preferably packaged as a bulk solution containing multiple doses in a pump spray system comprising a sealed container fitted with a metering pump.
In certain alternate embodiments the formulations according to the invention are preferably package as a single unit dose solution in a single unit dose pump spray system comprising a sealed container fitted with a pump.
Typically a patient is treated by administration sublingually of 1 to 2 actuations, from the spray pump. Another advantage of sublingual spray delivery is the ability to easily titrate patients by 1 or 2 doses as required by a single actuation. This is typically not the case with other forms of drug delivery (patches, lozenges, tablets, suppositories).
Pump action sprays are characterized in requiring the application of external pressure for actuation, for example, external manual, mechanical or electrically initiated pressure. This is in contrast to pressurized systems, e.g., propellant-driven aerosol sprays, where actuation is typically achieved by controlled release of pressure e.g., by controlled opening of a valve.
In certain embodiments the pump sprays are preferred as the use of a pump spray with the formulation of the present invention allows for the administration of droplets or particles having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns, and/or preferably having a size distribution of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns. This is in contrast to a pressurized system which may result in particles less than 5 microns. Liquid droplets or particles having a diameter of less than about 5 microns have the potential to enter into the lungs of a human upon administration. Such entry into the lungs could lead to an increase in patient to patient variability in absorption of the fentanyl. Further, absorption of fentanyl in the lungs could lead to an increased absorption and increased side effects, including respiratory depression which may be fatal.
In certain preferred embodiments, the droplet size of the delivered formulations further provides for an increase in surface area by being sprayed sublingually as opposed to being placed under the tongue with e.g., a dropper.
In certain preferred embodiments, the delivery device is a device such as those described in U.S. Pat. Nos. 6,866,566; 6,877,672; 6,772,915; 6,725,857; 6,705,493; 6,679,248; 6,578,741; 6,527,144; 6,484,715; 6,478,196; 6,461,322; 6,446,839; 6,427,878; 6,367.473; 6,364,166; 6,321,942; 6,234,366; 6,227,413; 6,059,151; 6,059,150; 6,055,979; 5,944,222; 5,901,883; 5,813,570; 4,565,302; 4,532,967; 6,964,381; 6,860,411; 6,824,020; 6,817,490; 6,585,172; 6,443,370; 6,427,680; 6,425,499; 6,401,987; 6,398,074; 6,264,065; 5,950,877; 5,328,099; 5,301,846; and the like which are described in certain embodiments as being suitable for nasal administration.
Other devices suitable for use in accordance with the formulations of the present invention are described in U.S. Pat. Nos. 6,808,085; 6,736,293; 6,732,955; 6,708,846; 6,626,379; 6,626,330; 6,626,328; 6,454,185; 6,427,876; 6,427,684; 6,419,167; 6,405,903; 6,352,181; 6,308,867; 6,257,461; 6,257,454; 6,250,509; 6,227,415; 6,209,760; 6,179,164; 6,109,547; 6,062,430; 6,026,992; 5,992,704; 5,992,703; 5,988,449; 5,967,369; 5,964,417; 5,950,879; 5,938,125; 5,927,559; 5,921,444; 5,893,484; 5,875,938; 5,862,962; 5,860,567; 5,816,504; 5,813,570; 5,803,311; 5,791,518, 5,692,650; 5,655,689; 5,584,417; 5,520,337; 5,519,980; 5,482,193; 5,469,989; 5,443,185; 5,439,177; 5,437,398; 5,427,280; 5,395,032; 5,375,745; 5,368,201; 5,366,122; 5,366,122; 5,335,823; 5,326,000; 5,323,936; 5,316,198; 5,301,841; 5,295,628; 5,289,946; 5,277,334; 5,257,726; 5,228,586; 5,209,375; 5,203,840; 5,147,087; 5,115,980; 5,110,052; 5,011,046; 4,958,752; 4,946,069; 4,944,430; 4,934,568; 4,921,142; 4,871,092; 4,830,284; 4,826,048; 4,823,991; 4,821,923; 4,817,829; 4,776,498; 4,762,475; 4,728,008; 4,726,747; 4,694,977; 4,694,976; 4,566,611; 6,851,583; 6,824,021; 6,779,690; 6,776,312; 6,971,559; 6,948,640; 6,945,473; 6,938,802; 6,933,850; 6,929,156; 6,918,514; 6,913,205; 6,866,168; 6,832,072; 6,830,163; 6,817,490; 6,817,489; 6,811,060; 6,811,057; 6,805,301; 6,805,263; 6,789,750; 6,789,706; 6,786,369; 6,783,035; 6,772,913; 6,769,579; 6,758,371; 6,752,298; 6,742,677; 6,705,062; 6,698,627; 6,698,623; 6,663,019; 6,659,314; 6,659,307; 6,655,550; 6,655,549; 6,651,846; 6,601,735; 6,595,395; 6,592,010; 6,588,629; 6,581,852; 6,571,991; 6,554,160; 6,536,635; 6,527,149; 6,527,148; 6,488,185; 6,471,097; 6,460,781; 6,460,740; 6,460,738; 6,446,841; 6,422,429; 6,409,049; 6,398,079; 6,360,919; 6,349,856; 6,345,737; 6,343,722; 6,662,561; 6,315,169; 6,273,303; 6,273,300; 6,261,274; 6,257,457; 6,234,363; 6,234,168; 6,221,054; 6,209,759; 6,189,741; 6,186,371; 6,155,496; 6,119,897; 6,105,826; 6,021,930; 6,012,615; 5,988,496; 5,950,871; 5,931,386; 5,850,948; 5,803,318; 5,799,810; 5,769,325; RE35,683; 5,692,492; 5,568,884; 5,566,865; 5,511,698; 5,482,188; 5,476,198; 5,366,115; 5,337,923; 5,249,713; 5,237,797; 5,234,135; 5,226,563; 5,190,192; 5,176,296; 5,127,548; 4,966,313; 4,91,840; 4,245,967; 4,030,667; and the like.
All of the patents recited herein are hereby incorporated by reference in their entireties. Although the delivery devices disclosed in the patents described above may be suitable for nasal or inhalation administration, in accordance with certain embodiments of the present invention the delivery devices are specifically adapted to be suitable for sublingual administration of a liquid formulation. In certain embodiments, the devices utilized to practice the present invention include components made from Pfeiffer of America, Inc., for example, Pfeiffer of America, Inc. sublingual unit dose device article reference number 72772. In other embodiments, the device is Pfeiffer of America, Inc., sublingual unit dose applicator assembly.
Preferably the device in accordance with the present invention is adapted to sublingually deliver the sublingual formulation in a controlled manner preferably such that only droplets having a mean diameter of at least about 10 microns, preferably at least about 20 microns, more preferably a mean diameter of from about 20 to about 200 microns are delivered to the patient. More preferably only droplets having a size distribution in the range of from about 5 microns to about 500 microns, preferably from about 10 microns to about 200 microns, preferably from about 20 microns to about 100 microns, more preferably from about 30 microns to about 70 microns.
Preferably the dispenser is constructed in such a way that it can be carried and simultaneously reliably operated with the fingers, or with three fingers of one hand, and can be used, for example, in the manner of a sublingual spray. The dispenser can be constructed as a disposable dispenser which, following the emptying of the medium chamber, does not have to be refilled and can therefore be constructed as a simple standard component, which receives the pump, the formulation, the channels and optionally, valves or closures within an outer casing, which in side view can be roughly T-shaped or Y-shaped.
If the dispenser is to be emptied in a single pump stroke in successive portions or in one complete pump stroke, and is not to be refilled, then the dispenser can be substantially tightly closed with respect to the outside in the starting position
In certain preferred embodiments, the delivery device (e.g., such as a spray pump device) includes a lock-out mechanism. Preferably the lock-out mechanism allows for administration of only one unit dose, and preferably prevents abuse of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, by only allowing for the administration of one dose and locking out of further administration for a certain and/or predetermined period of time. In certain embodiments, after one or more actuating cycles the actuator can be automatically transferred into the locking position, so that for performing a following actuating cycle randomly or deliberately a release must take place. Locking can take place in the starting position, actuating position and/or an intermediate position and can act both against actuation and against return or against one of these movements alone and several locking positions with the same or different locking action are possible.
In certain embodiments, the device may be premetered or alternatively, the device may be device-metered. Premetered devices preferably contain previously measured doses or a dose fraction in some type of units (e.g., single unit dose amount of solution, single or multiple blisters or other cavities) that may be included in the device during manufacture or by the patient before use. Typical device-metered units have a reservoir containing formulation sufficient for multiple doses that are delivered as metered sprays by the device itself when activated by the patient.
Important factors to consider with manufacture of the device are the reproducibility of the dose, the spray plume, and the particle/droplet size distribution, which can affect the delivery of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, under the tongue. Maintaining the reproducibility of these parameters through the expiration dating period and ensuring the functionality of the device (e.g., spray mechanism, electronic features, sensors) through its lifetime under patient-use conditions is important as any alteration in these parameters could lead to variability in dosing and absorption, which could lead to potential side effects.
The administered dose of spray drug formulation may be dependent on the design, reproducibility, and performance characteristics of the container closure system. A suitable device which provides the desired droplet/particle size distribution is an important factor for the correct performance of the fentanyl product. Actuation parameters (e.g., force, speed, hold and return times) should also be considered with respect to the device. Moreover, the device should be compatible with formulation components. Further, the device should be designed to prevent partial metering of the fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, formulation when used according to the patient instructions for use.
A typical device includes a base unit, a discharge actuator, an orifice for the formulation to be release from the device, and a medium reservoir. Preferably a reservoir is provided which as a dispensing chamber is filled already on production of the device. The medium reservoir preferably defines a measured content of fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, to be discharged upon a single activation.
In accordance with certain embodiments of the invention, a reservoir, or a space thereof receiving the medium is preferably an elongated shape preferably having a wall thickness which is constant over the circumference and length of the reservoir body. The reservoir body may be formed simply by a section of a cylindrical hollow of a plastics, steel, such as stainless steel, transparent material, such as glass, or the like so that its production is very simple.
Preferably an actuator body is provided on a unit of the device, which is movable relative to the orifice for activating discharge. This body, in the course of the actuating movement, opens a closure of a chamber, e.g. by puncturing. The space within this chamber may directly adjoin the medium in the reservoir, accommodate the opening body or the reservoir at least in part and configured as a pressure space which prior to being opened is at an elevated pressure. The opening body may be formed directly by the reservoir.
Preferably during a part of the actuating travel following the starting position an elevated pressure is built up. In a subsequent portion of the actuating movement continuing in the same direction, the medium is relieved of the pressure at one of the sides and communicated to the medium orifice on this side. As such, due to the pressure acting on the side, the medium is pushed from the reservoir and through the orifice.
Typically as the liquid formulation leaves the orifice, the liquid droplets follow a trajectory which is influenced by the orifice shape of the device. In certain embodiments, the droplet size, spray geometry, and the spray pattern are dependent on the design of the pump and/or the properties of the formulation. In certain embodiments, the orientation of the actuator, pump design, and the properties of the formulation will influence the spray symmetry and the shape.
In certain embodiments, the device of the present invention further includes a stopper. Preferably the stopper comprises a material which precludes or substantially precludes the absorption of the fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof. A suitable stopper for use in accordance with the device of the present invention is, for example, a stopper marketed by West Pharmaceutical Services, Inc. In certain preferred embodiments, the stopper has the following composition and characteristic: 1) elastomer: bromobutyl and/or chlorobutyl; 2) reinforcement: inert material; and 3) curing system: unconventional.
In certain embodiments, the device further includes a gasket. Preferably the gasket comprises a material which precludes or substantially precludes the absorption of the fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof. A suitable gasket for use in accordance with the device of the present invention is, for example, a stopper marketed by West Pharmaceutical Services, Inc. In certain preferred embodiments, the gasket has the following composition and characteristic: 1) elastomer: bromobutyl and/or chlorobutyl; 2) reinforcement: inert material; and 3) curing system: unconventional.
Droplet size distribution can be determined by utilizing any reliable method known to one of skill in the art. One such method uses laser diffraction devices, such as, for example, the Malvern Spraytec® with RT Sizer software. A Malvern Mastersizer S, by Malvern Instruments Limited (U.K.), device may also be used to determine size distribution. A Malvern Mastersize S is a modular particle size analyzer offering measurement versatility. It can measure spray droplet size as well as wet and dry samples. Particles from sub-micron to a few millimeters may be measured with the Malvern Mastersizer S.
Further, automated actuation stations for comparative in vitro bioequivalence tests or other testing to decrease the variability associated with manual actuation may also be used when determining the droplet size distribution. Any such automated actuation stations known to one of skill may be applicable in practicing the present invention. An example of one such device is the MightyRunt Actuation Station by Innova Systems, Inc. In a preferred embodiment, a MightyRunt is equipped with an exhaust fan attachment. In a further embodiment, the MightyRunt is further equipped with a Mettler Toledo balance Model AT201.
Other Active Agents
Although the invention described herein has been described with respect to fentanyl, a pharmaceutically acceptable salt thereof, or derivative thereof, it is contemplated that other active pharmaceutical agents, particularly those suitable for sublingual administration, may be used in accordance with the present invention. For example, in certain embodiments, other opioid analgesics which are suitable for sublingual administration may be used in place of the fentanyl, pharmaceutically acceptable salt thereof, or derivative thereof. Certain opioid analgesics for use in accordance with the present invention include, for example and without limitation, alfentanil, sufentanil, buprenorphine, butorphanol, codeine, hydrocodone, hydromorphone, levorphanol, meperidine, methadone, morphine, nalbuphine, oxycodone, oxymorphone, propoxyphene, tramadol, and the like.
In certain embodiments the active compound is an analgesic, opioid antagonist, anti-migraine agent, anti-emetic agent, anti-epileptic agent, anti-hypertensive agent, anesthetic agent, cannabinoid, cannabinoid antagonist, inverse agonist of cannabinoid, endocannabinoid, enkephalin, analogues or derivatives thereof, or mixtures thereof.
Analgesics useful in the present invention include, but are not limited to, alfentanil, buprenorphine, butorphanol, codeine, fenpipramide, hydrocodone, hydromorphone, levorphanol, meperidine, methadone, morphine, nalbuphine, oxycodone, oxymorphone, pentazocine, piritramide, propoxyphene, sufentanil, tilidine, tramadol, analogues or derivatives thereof, or mixtures thereof.
Opioid antagonists useful in the present invention include, but are not limited to, naloxone, naltrexone, nalmefene, analogues or derivatives thereof, or mixtures thereof.
Anti-migraine agents useful in the present invention include, but are not limited to, almotriptan, eletriptan, frovatriptan, naratriptan, rizatriptan, sumatriptan, zolmitriptan, ergot alkaloids, proxibarbal, lisuride, methysergide, clonidine, pizotifene, analogues or derivatives thereof, and mixtures thereof.
Anti-emetic agents useful in the present invention include, but are not limited to bromopride, domperidone, granisetron, ondansetron, tropisetron, metoclopramide, pyridoxine, scopolamine, thiethylperazine, analogues or derivatives thereof, or mixtures thereof.
Anti-epileptic agents useful in the present invention include, but are not limited to barbiturates, carbamazepine, ethosuximide, mesuximide, phenyloin, primidone, sultiam, valproic acid, vigabatrine, analogues or derivatives thereof, or mixtures thereof.
Anti-hypertensive agents useful in the present invention include, but are not limited to diltiazem, clonidine, nifedipine, verapamil, isosorbide-5-mononitrate, organic nitrates, agents used in treatment of heart disorders, analogues or derivatives thereof, or mixtures thereof.
Anesthetics useful in the present invention include, but are not limited to benzocaine, bupivacaine, dibucaine, etidocaine, levobupivacaine, lidocaine, mepivacaine, oxybuprocaine, piperocaine, prilocaine, procaine, proparacaine, ropivacaine, tetracaine, xylocaine, desflurane, enflurane, isoflurane, sevoflurane, benzonatate, dyclonine, ketamine, phenol, propofol, analogues or derivatives thereof, or mixtures thereof.
Cannabinoids useful in the present invention include, but are not limited to delta-8-tetrahydrocannabinol, delta-9-tetrahydrocannabinol, cannabidol, olivetol, cannabinol, cannabigerol, nabilone, delta-9-tetrahydro cannabinotic acid, the non-psychotropic cannabinoid 3-dimethylnepty 11 carboxylic acid homologine 8, delta-8-tetrahydrocannabinol, pharmaceutically acceptable salts thereof, complexes thereof, derivatives thereof, or mixtures thereof. A particularly preferred cannabinoid is delta-9-tetrahydrocannabinol, also known as dronabinol.
Further, active agents having a narrow therapeutic index or range (e.g., wherein small variances in blood levels of the drug causes changes in the effectiveness or toxicity of that drug) could be particularly suitable for use in accordance with the present invention. Such active agents include for example and without limitation, digoxin, levothyroxine, aminoglycosides (e.g., gentamycin, tobramycin), antiarrythimics (e.g., procainamide, quinidine), theophylline, antineoplastics, busulfan, methotrexate, 6-MP, carboplatinum, antidepressants (e.g., lithium), anticonvulsants (e.g., phenyloin, carbamazepine, valproate sodium, valproic acid), antipsychotics, anticoagulants (e.g., warfarin), cyclosporine, and the like.
The present invention will now be more fully described with reference to the accompanying examples. It should be understood, however, that the following description is illustrative only and should not be taken in any way as a restriction on the generality of the invention specified above.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS Example 1
In Example 1, a fentanyl sublingual formulation was prepared having a concentration of 0.5 mg/ml. The formulation is listed in Table 1 below:
TABLE 1
Ingredient Percent
Concentration of Fentanyl percent to make 0.5 mg/ml
Base
Ethanol % (v) 20
Propylene glycol % (v)  5
DI Water % (v) QS
Example 2
In Example 2, a fentanyl sublingual formulation was prepared having a concentration of 0.5 mg/ml. The formulation is listed in Table 2 below:
TABLE 2
Ingredient Percent
Concentration of Fentanyl percent to make 0.5 mg/ml
Base*
Ethanol % (v) 20
Propylene glycol % (v)  5
DI Water % (v) QS
Contains fentanyl citrate equivalent to 0.5 mg/ml of fentanyl base
Example 3
In Example 3, a fentanyl sublingual formulation was prepared having a concentration of 0.5 mg/ml. The formulation is listed in Table 3 below:
TABLE 3
Ingredient Percent
Concentration of Fentanyl percent to make 0.5 mg/ml
Base
Ethanol % (v) 20
Propylene glycol % (v) 5
DI Water % (v) QS
Mannitol % (wt) 0.3
Tween 80 % (wt) 0.2
Example 4
In Example 4, a fentanyl sublingual formulation was prepared having a concentration of 0.5 mg/ml. The formulation is listed in Table 4 below:
TABLE 4
Ingredient Percent
Concentration of Fentanyl percent to make 0.5 mg/ml
Base
Ethanol % (v) 20
Propylene glycol % (v) 5
Buffer % (v) QS
Triacetin % (wt) 0.5
Example 5
In Example 5, a fentanyl sublingual formulation was prepared having a concentration of 0.5 mg/ml. The formulation is listed in Table 5 below:
TABLE 5
Ingredient Percent
Concentration of Fentanyl percent to make 0.5 mg/ml
Base
Ethanol % (v) QS
Propylene glycol % (v) 5
Miglyol % (v) 50%

Preparation of Formulations (Examples 1-5)
  • 1. Calculated amount of Fentanyl base or Fentanyl citrate was weighed in a tared glass container.
  • 2. Calculated amount of alcohol was added to the container and mixed to dissolve fentanyl.
  • 3. Propylene glycol was weighed and added to the fentanyl solution.
  • 4. Water or Buffer or Miglyol was weighed, added to the fentanyl solution and mixed for 2 mm.
  • 5. Inactive ingredients (Mannitol, Triacetin, or TW80) were added at the end and mixed well.
  • 6. The final solution was vortexed for 3 min. After mixing, the formulations were stored in refrigerator for further studies.
Example 6
In Example 6, three rabbits weighing 2-3 kg were used to study the bioavailability of sublingual administration of Examples 1-5 in comparison to the IV injection of the formulation as a reference. Rabbits were first anesthetized by Isoflurane gas as needed to keep the rabbits immobilized for approximately 15-20 min.
For each formulation, study rabbits received a single dose of 0.1 ml (equivalent to 50 μg of fentanyl base) by sublingual and IV administration. For the sublingual studies, the dose of liquid formulation was administered underneath the tongue using a spray bottle. Blood samples (1 ml per sample) were obtained through a catheter installed inside the ear vein.
Blood samples were collected at zero time baselines and at 5, 10, 20, 45, 60 and 120 min after the single dose. Samples were immediately cooled and plasma was separated by centrifugation within 2-3 hrs of blood collection. Samples were stored at −20° C. until assayed. After recovering from anesthesia, animals were returned to their cages. These animals were rested in cages for at least 5-7 days before they could be reused for further testing.
Plasma Collection & Separation
Blood was collected from rabbits in 3 ml tubes containing 7.5% EDTA and centrifuged at 3,000 RPM for 15 min to remove blood cells and other insoluble material. The plasma was decanted into a silanized tube and kept frozen at −20° C. until assay.
Plasma Extraction
For each plasma sample (0.5 ml), 100 μl of Sufentanil (IS) and 100 μL 5M NaOH (for protein denaturation) were added. The Fentanyl was extracted with 1 ml of 1-chlorobutane by vortex mixing for 15 min. After centrifuging at 12000 RPM for 5 min and freeze-drying for 10 min (to break up any emulsion), the upper organic layer was decanted and evaporated to dryness using a gentle stream of nitrogen. The extraction residue was reconstituted with 100 μl methanol followed by vortex mixing for 5 min and sonicating for 3 min, then a 3 μl volume was injected into the GC-MS system.
Results and Discussion
Plasma Concentration vs. Time Profiles
Plasma concentration-time profiles of Fentanyl in rabbits following IV and sublingual administration of 0.1 ml (equivalent to 50 pg of fentanyl base) are given in the following tables (Tables 6A-10B).
Tables 6A and 6B provide the plasma concentration-time profiles for Example 1.
TABLE 6A
(Example 1 intravenous)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 785.8012 839.9696 812.885396 38.302813
10 507.7546 715.6065 611.680527 146.97351
20 737.3449 423.5903 667.9939 609.643027 164.81539
45 701.5631 482.215 382.5801 522.119422 163.19256
60 562.501 554.8479 475.4686 530.939148 48.191101
90 207.5541 492.4037 429.1217 376.359829 149.57498
120 214.0297 196.9675 205.4986 12.064797
TABLE 6B
(Example 1 sublingual)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 412.1095 370.7992 391.454361 29.210825
10 205.5274 279.3298 755.3469 413.401352 52.186202
20 539.2677 627.0507 712.6511 626.323191 86.693973
45 618.9493 387.7606 503.35497 163.47506
60 349.3563 218.14 498.3773 355.291188 140.2129
90 245.8519 249.5091 231.7688 242.376606 9.3668848
120 214.1339 162.9939 199.6146 192.247465 26.353936
In Tables 7A and 7B are the plasma concentration-time profiles for Example 2.
TABLE 7A
(Example 2 intravenous)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 671.5152 788.2252 729.870183 58.35497
10 379.1866 617.5517 1102 699.579446 300.73378
20 603.9696 407.5375 505.75355 98.216024
45 380.9878 292.998 336.992901 43.994929
60 140.7566 266.3611 388.5314 265.216362 101.1569
90 81.06491 254.4848 334.8012 223.450304 105.88636
120 95.34888 278.7789 232.4037 202.177147 77.875432
TABLE 7B
(Example 2 sublingual)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 350.2297 106.9249 337.6856 264.946755 136.99455
10 373.5822 115.1643 486.0254 324.923935 190.15835
20 285.5994 294.5517 518.5091 366.220081 131.96212
45 302.7099 52.78093 359.9483 238.479716 163.34652
60 118.3915 43.02434 314.9564 158.790737 140.39528
90 81.06491 43.39148 62.2281947 26.639136
120 95.34888 35.34888 30.68154 53.7931034 36.063946
In Tables 8A and 8B are the plasma concentration-time profiles for Example 3.
TABLE 8A
(Example 3 intravenous)
TIME (min) R1 (ng/ml) R2 (ng/ml) R3 (ng/ml) GRAND AVG STD DEV
5 903.4949 1000.205 882.0649 928.588235 51.390778
10 814.3834 557.9432 484.5233 618.949966 141.40552
20 348.8641 309.1785 329.021298 19.842799
45 120.2677 169.7972 211.0669 167.043949 37.119701
60 160.3347 121.5882 128.9087 136.943881 16.807631
90 89.85598 85.70081 92.71197 89.4229209 2.8786265
120
TABLE 8B
(Example 3 sublingual)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 127.3124 269.1927 198.252535 100.32454
10 304.6288 589.1684 310.5953 401.464165 162.58397
20 349.3611 689.7999 281.5365 440.232477 218.77603
45 288.0639 418.1555 195.9391 300.719518 111.64744
60 173.0345 255.9016 214.45 74.967507
90 224.432 87.7931 228.6156 180.280257 80.123534
120 96.5284 145.1907 120.859533 34.409422
In Tables 9A and 9B are the plasma concentration-time profiles for Example 4.
TABLE 9A
Example 4 intravenous)
TIME R1 R2 R3
(min) (ng/ml) (ng/ml) (ng/ml) GRAND AVG STD DEV
5 740.3759 732.5855 717.8438 730.268425 11.443374
10 666.2644 671.4571 627.0568 654.926076 24.274753
20 713.355 592.5 638.7444 648.199797 60.979784
45 575.2667 557.7789 482.8316 538.625761 49.103907
60 409.8756 596.6278 548.1217 518.208362 96.903054
90 455.8567 484.928 430.5538 457.112801 27.208875
120 385.2982 452.6998 418.998986 47.660144
TABLE 9B
(Example 4 sublingual)
TIME R1 R2 R3 GRAND
(min) (ng/ml) (ng/ml) (ng/ml) AVG STD DEV
5 250.8854 481.6146 459.2779 397.259297 127.25455
10 478.074 577.9594 614.8479 556.960446 70.76361
20 587.8195 465.9229 518.4341 524.058824 61.142626
45 538.3245 499.217 449.574 495.705206 44.479353
60 367.4361 452.9817 442.7566 421.058147 46.718636
90 309.5538 571.0487 202.8722 361.158215 189.43533
120 205.6531 195.7282 427.1927 276.191346 130.86513
In Tables 10A and 10B are the plasma concentration-time profiles for Example 5.
TABLE 10A
(Example 5 intravenous)
TIME (min) R1 (ng/ml) R2 (ng/ml) R3 (ng/ml) GRAND AVG STD DEV
5 928.5193 1247.247 1123.335 1099.70047 160.67311
10 920.1521 1100.3 1103.844 1041.43205 105.04643
20 876.3793 966.998 972.8114 938.729547 54.075067
45 765.9696 938.3834 947.0609 883.804598 102.14032
60 645.6045 892.6836 482.2799 673.52265 206.62129
90 157.9533 418.6034 343.0811 306.545977 134.1109
120 58.43813 30.68154 30.68154 39.933739 16.025276
TABLE 10B
(Example 5 sublingual)
TIME (min) R1 (ng/ml) R2 (ng/ml) R3 (ng/ml) GRAND AVG STD DEV
5 140.7911 176.7338 127.357 148.293949 25.529127
10 239.712 191.2956 210.8458 213.951149 24.357082
20 409.5335 580.6095 540.5842 510.242394 89.483087
45 351.3955 500.9315 314.7343 389.020453 98.636103
60 364.9493 439.2885 280.645 361.62762 79.373899
90 245.2698 403.4255 30.68154 226.458925 187.08258
120 30.68154 119.7079 215.6552 122.014875 92.508392

Pharmacokinetic Parameters
The pharmacokinetic parameters Peak plasma concentration (Cmax), Time to reach Cmax (Tmax), Half-life (t1/2), Area under the curve (AUC), and Total body clearance (CL) obtained after sublingual or IV drug administrations are tabulated in the following tables (Tables 11-15). Also, plasma concentration-time curves after administering IV and sublingual doses of Fentanyl are shown in FIGS. 1-10.
TABLE 11
(Example 1 intravenous and sublingual)
ROUTE OF AUC T1/2 TMAX CMAX VD Cl
ADMINISTRATION (min * ng/ml) (min) (min) (ng/ml) (ml) (ml/min)
IV 71772.86 43.8141 0 812.8854 50.7843 0.6966
SL 59684.34 55.704 20 626.3232
TABLE 12
(Example 2 intravenous and sublingual)
ROUTE OF AUC T1/2 TMAX CMAX VD Cl
ADMINISTRATION (min * ng/ml) (min) (min) (ng/ml) (ml) (ml/min)
IV 86790.18 153.2372 0 729.8702 181.8356 0.9218
SL 23759.14 33.1477 20 366.2201
TABLE 13
(Example 3 intravenous and sublingual)
ROUTE OF AUC T1/2 TMAX CMAX VD Cl
ADMINISTRATION (min * ng/ml) (min) (min) (ng/ml) (ml) (ml/min)
IV 32707.76 49.7523 0 928.5882 78.4776 1.5287
SL 40623.13 57.6038 20 440.2325
TABLE 14
(Example 4 intravenous and sublingual)
ROUTE OF AUC T1/2 TMAX CMAX VD Cl
ADMINISTRATION (min * ng/ml) (min) (min) (ng/ml) (ml) (ml/min)
IV 185485.6 200.3556 0 730.2684 77.036 0.2696
SL 86916.91 93.4018 10 556.9604
TABLE 15
(Example 5 intravenous and sublingual)
ROUTE OF AUC T1/2 TMAX CMAX VD Cl
ADMINISTRATION (min * ng/ml) (min) (min) (ng/ml) (ml) (ml/min)
IV 76250.66 17.0944 0 1099.7005 27.3544 0.6557
SL 42554.59 38.2788 20 510.2424
The maximum concentrations were reached in about 20 minutes for all formulations after sublingual administration. There was considerable inter-individual variability by both routes of administration. Measurable plasma concentration after sublingual administration was demonstrable after 120 min in most formulation testing. In conclusion, sublingual fentanyl administration showed good absorption profile compared to IV.
Example 7
In Example 7, a study was conducted to determine the pharmacokinetics of a formulation prepared in accordance with Example 1 after increasing sublingual dose administration in healthy volunteers under fasting conditions. The study was also conducted to determine the safety and tolerability of a fentanyl sublingual spray prepared in accordance with Example 1 in humans.
The study was a single center, single dose, single-blinded, sequential ascending dose and repeated design in healthy male subjects. The following treatments were to be administered under fasting conditions:
Treatment A (Test 1): One Fentanyl 1 mg/ml sublingual spray (1×100 μg dose)
Treatment B (Test 2): One Fentanyl 4 mg/ml sublingual spray (1×400 μg dose)
Treatment C (Test 2): Two Fentanyl 4 mg/ml sublingual spray (1×800 pg dose)
Treatment D (Placebo-Test 1): One Placebo 1 mg/ml sublingual spray (1×100 μg dose)
Treatment E (Placebo-Test 2): One Placebo 4 mg/ml sublingual spray (1×400 μg dose)
Treatment F (Placebo-Test 2): Two Placebo 4 mg/ml sublingual spray (1×800 μg dose)
The products were to be administered to nine (9) healthy male volunteers according to the following design in Table 16 below:
TABLE 16
Period 1 Period 2 Period 3
Sequence 1 (n = 6) 100 μg 400 μg 800 μg
Sequence 2 (n = 3) Placebo Placebo Placebo
In each period, subjects were to arrive at the clinical site at least 10 hours before dosing. After a supervised overnight fast, a single oral dose of the assigned formulation was to be orally administered in the morning. Subjects were allowed to leave the clinical site after the 24-hour blood draw. The wash-out period was to be of at least 14 days; the duration of this study was expected to be approximately 5-6 weeks. As per protocol, each sequential dose was to be separated by a wash-out of at least 14 days, which corresponds to more than 10 time the expected half life of the moiety to be measured. However, during the study periods 1 and 2 were separated by a wash-out of 15 days and periods of 2 and 3 were separated by a wash-out of 13 days. As fentanyl's expected half life is reported to be approximately 6.4 hours, it is judged that these wash-out deviations should not affect the conclusion of the study. Furthermore, no carry-over was observed at the beginning of the second and third periods.
Pharmacokinetic Assessments
Blood samples for pharmacokinetic measurements were collected prior to and up to 24 hours (serial samplings) after each drug administration. The drug concentrations produced by the administration of the studied formulations were used to derive the pharmacokinetic parameters listed in Tables 17 and 18 below.
Six (6) subjects were included in the statistical analysis. A summary of the non-normalized pharmacokinetic parameters is presented in Table 17 and a summary of the normalized pharmacokinetic parameters is presented in Table 18. The mean measured plasma concentrations versus time profile, produced by the administration of the Test products, is depicted in FIG. 11, whereas the In-transformed mean concentrations versus time profile is presented in FIG. 12.
The pharmacokinetic parameters of interest for this study were Cmax, AUC, AUCT, AUCT/∞, Kel, Tmax, T1/2el, Cl/F and VA/F.
TABLE 17
Pharmacokinetic Parameters
Fentanyl (n = 6)
Non-normalized Data
TEST 1 (100 μg) TEST 2 (400 μg) TEST 2 (800 μg)
n = 6 n = 6 N = 2
PARAMETER MEAN C.V. (%) MEAN C.V. (%) MEAN C.V. (%)
Cmax (pg/ml) 172.0 27.1 708.0 50.2 1270.4 37.7
ln (Cmax) (pg/ml) 5.1207 4.8 6.4509 8.2 7.1102 5.4
Tmax (hours) 0.50 29.7 0.50 61.3 0.75 0.0
AUCT (pg · h/ml) 472.6 66.2 3556.1 63.0 5417.3 30.6
ln (AUCT) (pg · h/ml) 6.0271 8.6 8.0208 7.5 8.5734 3.6
AUC(pg · h/ml) 817.9 36.1 4242.6 57.6 5726.8 28.8
ln (AUC) (pg · h/ml) 6.6607 4.8 8.2303 6.4 8.6317 3.4
AUCT/∞ (%) 54.90 28.7 81.48 10.6 94.35 1.8
Kel (hour−1) 0.2008 27.4 0.1593 44.9 0.1782 0.9
T1/2el (hours) 3.70 30.4 5.20 45.8 3.89 0.9
Cl/F (ml/h/kg) 1718.8 27.7 1532.2 49.7 1837.3 13.1
Vz/F (ml/kg) 9070.2 34.7 10470.4 47.1 10307.7 12.2
For Tmax, the median is presented and the statistical analysis is based on ranks.
TABLE 18
Data Normalized to the 100 μg dose
TEST 1 (100 μg) TEST 2 (400 μg) TEST 2 (800 μg)
n = 6 n = 6 N = 2
PARAMETER MEAN C.V. (%) MEAN C.V. (%) MEAN C.V. (%)
Cmax (pg/ml) 172.0 27.1 177.0 50.2 158.8 37.7
ln (Cmax) (pg/ml) 5.1207 4.8 5.0646 10.4 5.0307 7.7
Tmax (hours) 0.50 29.7 0.50 61.3 0.75 0.0
AUCT (pg · h/ml) 472.6 66.2 889.01 63.0 677.2 30.6
ln (AUCT) (pg · h/ml) 6.0271 8.6 6.63461 9.1 6.4940 4.8
AUC(pg · h/ml) 817.9 36.1 1060.7 57.6 715.9 28.8
ln (AUC) (pg · h/ml) 6.6607 4.8 6.8440 7.7 6.5523 4.5
AUCT/∞ (%) 54.90 28.7 81.482 10.6 94.352 1.8
Kel (hour−1) 0.2008 27.4 0.1593 44.9 0.1782 0.9
T1/2el (hours) 3.70 30.4 5.20 45.8 3.89 0.9
Cl/F (ml/h/kg) 1718.8 27.7 1532.2 49.7 1837.3 13.1
Vz/F (ml/kg) 9070.2 34.7 10470.4 47.1 10307.7 12.2
For Tmax, the median is presented and the statistical analysis is based on ranks.
1= Different than Test-1 (p < 0.05)
2= Different than Test-1 (p < 0.01)
N.S. = Not Significant (p > 0.05)

Definition of the Pharmacokinetic Parameters
Cmax: Maximum plasma concentration (ng/ml).
Tmax: time of maximum measured plasma concentration; if it occurs at more than one time point, Tmax is defined as the first time point with this value (hour).
CLQC: Last quantifiable concentration (ng/ml).
TLQC: Time of last quantifiable plasma concentration (hour).
AUCT: Cumulative area Under the plasma concentration time Curve calculated from 0 to TLQC using the Trapezoidal method (ng·h/ml), and calculated according the following equation:
A U C T = 2 N TLQC [ ( C n - 1 + C n 2 ) × ( T n - T n - 1 ) ]
Where NTLQC is the number of the sample related to the TLQC.
AUC: Extrapolated Area Under the plasma concentration time Curve to infinity (ng·h/ml).
A U C = A U C T + ( C LQC K el )
AUCT/∞: Relative percentage of AUCT with respect to AUC(%).
A U C T / = ( A U C T A U C ) × 100
TLIN: Time point where log-linear elimination begins (hour).
Kel: Apparent mean elimination rate constant estimated by a non-linear least-squares regression analysis; a minimum of three values are required at the end of the concentration-time curve (hour−1).
T1/2el: Half-life of elimination (hour).
T 1 / 2 el = ln ( 2 ) K el
Cl/F: Apparent clearance (ml/h/kg)
Cl / F = Dose * AUC
VZ/F: Apparent volume of distribution (ml/kg)
V Z / F = Dose * K el · A U C * Dose expressed per weight ( kg )
As noted in Table 11, the parameters for which a statistically significant difference was observed between the 100 μg dose and the normalized 400 μg dose was AUCT, ln(AUCT) and AUCT/∞ and the parameter for which a statistically significant difference was observed between the 100 μg dose and the normalized 800 μg dose was AUCT/∞. No statistically significant differences were observed for any of the other pharmacokinetic parameters under study.
The mean Cmax value of the 100 μg dose was 172.0 pg/ml while for the 400 μg dose, it was 708.0 pg/ml and for the 800 μg dose, it was 1270.4 pg/ml. Once normalized to the 100 μg dose, the mean Cmax value of the 400 μg dose was 177.0 pg/ml and for the 800 pg dose, it was 158.8 pg/ml.
The median Tmax was 0.50 hour for the 100 μg dose and the 400 μg dose, and 0.75 hour for the 800 μg dose.
The mean AUCT value of the 100 pg dose was 472.6 pg·h/ml while for the 400 μg dose, it was 3556.1 pg·h/ml and for the 800 μg dose, it was 5417.3 pg·h/ml. Once normalized to the 100 μg dose, the mean AUCT value of the 400 μg dose was 889.0 pg·h/ml and for the 800 μg dose, it was 677.2 pg·h/ml.
The mean AUC value of the 100 g dose was 817.9 pg·h/ml while for the 400 μg dose, it was 4242.6 pg·h/ml and for the 800 μg dose, it was 5726.8 pg·h/ml. Once normalized to the 100 μg dose, the mean AUC value of the 400 pg dose was 1060.7 pg·h/ml and for the 800 μg dose, it was 715.9 pg·h/ml.
The AUCT/∞ ratio was approximately 55% for the 100 μg dose, 81% for the 400 μg dose and 94% for the 800 μg dose.
The mean Kel was 0.2008 hours−1 for the 100 μg dose, 0.1593 hours' for the 400 μg dose and 0.1782 hours' for the 800 μg dose, while the mean T1/2el value was 3.70 hours, 5.20 hours and 3.89 hours following the same order.
The mean Cl/F was 1718.8 ml/h/kg for the 100 μg dose, 1532.2 ml/h/kg for the 400 μg dose and 1837.3 ml/l/kg for the 800 μg dose, while the mean VZ/F was 9070.2 ml/kg for the 100 μg dose, was 10470.4 ml/kg for the 400 μg dose and was 10307.7 ml/kg for the 800 μg dose.
The intra-subject variation was 36.79%, 37.05% and 28.88% for Cmax, AUCT, and AUC, respectively.
Safety Evaluation
The safety parameters included the occurrence of adverse effects, measurements of vital signs, respiratory rate, oxygen saturation of blood by finger pulse oximetry, ECG and clinical laboratory parameters.
All adverse events were spontaneously reported by the volunteer, observed by the Clinical Investigator (or delegates) or elicited by general questioning by the clinical staff. Adverse events were also reported upon completion of the form “Taste of Medication Questionnaire to Subject”, which was filled right after dosing. For the purposes of the study, the period of observation for each individual subject extended from the time the subject gave informed consent to within 7 days following the last drug administration.
Safety Results:
All nine subjects experienced a total of one-hundred-twenty-seven (127) adverse events during the study. No serious adverse events were recorded in this study. Twenty adverse events (8 different types) were reported after the single dose administration of the Test 1 (A) product, fifty-six adverse events (26 different types) were reported after the single dose administration of the Test 2 (B) product, twenty-two adverse events (19 different types) were reported after the single dose administration of the Test 2 (C) product, ten adverse events (8 different types) were reported after the single dose administration of the Placebo-Test 1 (D) product, eleven adverse events (9 different types) were reported after the single dose administration of the Placebo-Test 2 (E) product and twelve adverse events (8 different types) were reported after the single dose administration of the Placebo-Test 2 (F) product. Two (2) adverse events associated with post-study laboratory test results were imputed to three formulations.
Six subjects (100%) reported adverse events after the administration of the Test 1 (A) formulation, six subjects (100%) reported adverse events after the administration of the Test 2 (B) formulation, two subjects (100%) reported adverse events after the administration of the Test 2 (C) formulation, three subjects (100%) reported adverse events after the administration of the Placebo-Test 1 (D) formulation, three subjects (100%) reported adverse events after the administration of the Placebo-Test 2 (E) formulation and three subjects (100%) reported adverse events after the administration of the Placebo-Test 2 (F) formulation. The adverse events by system Organ Class are listed in Table 19.
The events abdominal distension, abdominal pain, abdominal pain upper, anxiety, depressed mood, diarrhea, disturbance in attention, dizziness (10 episodes out of 11), dry mouth, dry skin, dysgeusia, headache, fatigue (6 episodes out of 7), feeling cold, feeling drunk, feeling hot, feeling of relaxation, hot flush, hyperhidrosis, hypoaesthesia oral, hypoaesthesia, nasal congestion, nausea, oral discomfort, pallor, paresthesia oral, pruritus, sensation of heaviness, somnolence, speech disorder, tongue coated and vomiting were assessed to be possibly related to the drugs. The other events cough, dizziness (1 episode out of 11), fatigue (1 episode out of 7), musculoskeletal pain, rhinorrhoea and throat irritation were assessed to be not related to the study drugs. The other event nasopharyngitis was assessed to be unlikely related to the study drugs.
TABLE 19
Summary of Adverse Events by System Organ Class
Period 1 Period 2 Period 3
Fentanyl Fentanyl Fentanyl
Parameter 100 μg Placebo 400 μg Placebo 800 μg Placebo
Number of subjects exposed 6 3 6 3 2 3
Number of subjects reporting 6 3 6 3 2 3
at least one adverse event
Total number of withdrawals 0 0 4 0 0 0
Withdrawals due to adverse 0 0 1 0 0 0
event (not related to tested drug)
Withdrawals due to adverse 0 0 2 0 0 0
event (related to tested drug)
Total number of adverse events* 20 10 56 11 22 12
Adverse events at least 18 8 53 11 22 12
possibly drug related*
Number % of Number % of Number % of Number % of Number % of Number % of
of subjects of subjects of subjects of subjects of subjects of subjects
Nature of adverse events subjects exposed subjects exposed subjects exposed subjects exposed subjects exposed subjects exposed
Gastrointestinal disorders
Abdominal distension 0 0 0 0 1 17 0 0 0 0 0 0
Abdominal pain 0 0 0 0 0 0 1 33 0 0 1 33
Abdominal pain upper 0 0 0 0 1 17 0 0 0 0 0 0
Diarrhoea 0 0 0 0 0 0 1 33 0 0 1 33
Dry mouth 0 0 0 0 0 0 0 0 1 50 0 0
Dysgeusia 6 100 3 100 6 100 3 100 2 100 3 100
Hypoaesthesia oral 4 67 1 33 3 50 1 33 1 50 0 0
Nausea 0 0 0 0 4 67 0 0 1 50 0 0
Oral discomfort 1 17 0 0 1 17 1 33 1 50 0 0
Tongue coated 0 0 0 0 1 17 0 0 0 0 0 0
Vomiting 0 0 0 0 1 17 0 0 1 50 0 0
General disorders and administration site conditions
Fatigue 3 50 1 33 2 33 0 0 1 50 0 0
Feeling cold 0 0 0 0 1 17 0 0 1 50 0 0
Feeling drunk 0 0 0 0 6 100 0 0 0 0 0 0
Feeling hot 0 0 0 0 1 17 0 0 1 50 0 0
Feeling of relaxation 3 50 0 0 1 17 0 0 0 0 0 0
Hyperhidrosis 0 0 0 0 0 0 0 0 1 50 0 0
Investigations*
Alanine aminotransferase 0 0 1 33 0 0 1 33 0 0 1 33
increased
Aspartate aminotransferase 0 0 1 33 0 0 1 33 0 0 1 33
increased
Musculoskeletal and connective tissue disorders
Sensation of heaviness 0 0 0 0 1 17 0 0 0 0 0 0
Musculoskeletal pain 0 0 0 0 1 17 0 0 0 0 0 0
Nervous system disorders
Dizziness 0 0 0 0 6 100 0 0 2 100 0 0
Disturbance in attention 0 0 0 0 2 33 0 0 1 50 0 0
Headache 0 0 0 0 1 17 0 0 0 0 1 33
Hypoaesthesia 0 0 0 0 4 67 0 0 1 50 0 0
Paresthesia oral 0 0 1 33 3 50 1 33 0 0 3 100
Somnolence 0 0 1 33 1 17 0 0 2 100 1 33
Speech disorder 0 0 0 0 0 0 0 0 1 50 0 0
Psychiatric disorders
Anxiety 0 0 0 0 0 0 1 33 0 0 0 0
Depressed mood 0 0 0 0 1 17 0 0 0 0 0 0
Respiratory, thoracic and mediastinal disorders
Cough 1 17 0 0 0 0 0 0 0 0 0 0
Nasal congestion 1 17 0 0 1 17 0 0 0 0 0 0
Nasopharyngitis 0 0 1 33 0 0 0 0 0 0 0 0
Rhinorrhoea 1 17 0 0 0 0 0 0 0 0 0 0
Throat irritation 0 0 0 0 1 17 0 0 0 0 0 0
Skin and subcutaneous tissue disorders
Dry skin 0 0 0 0 0 0 0 0 1 50 0 0
Pallor 0 0 0 0 1 17 0 0 1 50 0 0
Pruritus 0 0 0 0 0 0 0 0 1 50 0 0
Vascular disorders
Hot flush 0 0 0 0 3 50 0 0 1 50 0 0
*= Two (2) adverse events were associated with post-study laboratory test results and have been assigned to each period.

Conclusions
The pharmacokinetic parameters were well defined for the three doses (100 μg, 400 μg and 800 μg) administered in this study. Cmax and AUC seem to be proportional, AUCT is consistent with dose-proportionality between the 400 μg and 800 μg. Furthermore, the two formulations of fentanyl (1 mg/ml and 4 mg/ml sublingual spray in doses of 100 μg, 400 μg and 800 μg) administered during the study were well tolerated in most of the subjects. No subject participating in the trial reported serious adverse events during the course of this study.
Examples 8-12 In-vitro Permeation Experiments
The permeation characteristics of fentanyl formulations were studied using EpiOral tissues (ORL-100). MatTek Corp's EpiOral is used as model for screening sublingual drug absorption of pharmaceutical formulations. MatTek's EpiOral tissue consists of normal, human-derived epithelial cells. The cells have been cultured to form multilayered, highly differentiated models of the human buccal (EpiOral) phenotypes. The EpiOral tissue model exhibits in vivo-like morphological and growth characteristics which are uniform and highly reproducible. Morphologically, the tissue model closely parallels native human tissue, thus providing a useful in-vitro means to assess in-vivo permeability of pharmaceutical formulations across sublingual mucosa.
The EpiOral tissues, grown on cell culture inserts with Teflon backing membrane, were shipped by MatTek Corp on Monday for delivery on Tuesday morning. All the tissues were used in the permeability experiments within 72 hours of shipment. The inserts containing the tissues were rinsed with distilled water before the start of permeation experiments. The tissue area for the ORL-100 is 0.6 cm2.
The receiver compartment (wells) received 0.3 ml of phosphate citrate buffer of pH 6.6 (receiver solution). The donor compartments (tissue inserts) were placed in the wells and filled with 0.5 ml of drug solution (donor solution).
The inserts were moved from well to well containing fresh receiver fluid at pre-determined intervals (2, 5, 7, 9, 11, 13, 15, 30, 45, 60, 90, and 120 min). After permeation studies both donor and receiver fluids were collected in vials for analysis by HPLC. The receiver and donor solution concentrations and the flux over each permeation time interval were determined.
Example 8
In Example 8, the permeation of fentanyl base was compared to the permeation of fentanyl citrate. The formulations and percent permeated in 2 hours is listed in the table below:
TABLE 20
CONC. OF %
FENTANYL ALCOHOL PG MIGLYOL PERMEATED
EXAMPLE # * BASE % (V) % (V) % (V) IN 2 HOURS
Fentanyl 8-a (b) 1 mg/ml 20 5 17.33
Base
Fentanyl 8-b (w) 1 mg/ml 20 5 17.18
Base
Fentanyl 8-c (b) 0.646 mg/ml    20 5 1.81
Citrate
Fentanyl 8-d 1 mg/ml 20 5 79.3 1.64
Base
(b)—buffer,
(w)—water
Example 9
In Example 9, Effect of Alcohol on the Permeation of Fentanyl Base Formulations was tested. The formulations and percent permeated in 2 hours are listed in the Table below:
TABLE 21
CONC. OF % PERMEATED
FENTANYL ALCOHOL PG IN
EXAMPLE #* BASE % (V) % (V) 2 HOURS
9-a (w) 1 mg/ml 20 5 17.18
(Control)
9-b (w) 1 mg/ml 28 5 13.45
9-c (w) 1 mg/ml 40 5 10.95
9-c (w) 1 mg/ml 50 5 9.18
(b)—buffer,
(w)—water
Example 10
In Example 10, the effects of PG (propylene glycol) on Fentanyl Formulations was tested. The formulations and percent permeated in 2 hours are listed in the table below:
TABLE 22
CONC. OF
FENTANYL ALCOHOL PG % PERMEATED
EXAMPLE #* BASE % (V) % (V) IN 2 HOURS
10-a (w) 1 mg/ml 20 5 17.18
(Control)
10-b (w) 1 mg/ml 20 25 14.518
(b)—buffer,
(w)—water
Example 11
In Example 11, the effect of pH on Fentanyl Formulations was tested. The formulations and percent permeated in 2 hours are listed in the table below. As indicated in the table, the permeation of fentanyl across buccal tissue was dependent on the pH of the formulation. Because fentanyl has pKa value (7.3 and 8.4) within the pH range studied, its degree of ionization changed as the pH of the formulation was altered. The results in the table below indicate that that formulations adjusted to pH between 8 and 9 were showing better permeability and physical stability.
TABLE 23
CONC. OF %
FENTANYL BUFFER ALCOHOL PERMEATED
EXAMPLE #* BASE PH % (V) PG % (V) IN 2 HOURS
11-a (w) 2 mg/ml Water 30 5 17.83
(Control)
11-b (b) 2 mg/ml 5.5 30 4 5.33
11-c (b) 2 mg/ml 6.6 29 4 10.62
11-d (b) 2 mg/ml 8.6 29 4 13.48
11-e (b) 2 mg/ml 9.6 29 4 11.8
(b)—buffer,
(w)—water
Example 12
In Example 12, several ingredients including hydroxypropyl beta cyclodextrin (HPBCD), mannitol, polyvinyl pyrrolidone (PVP), propylene carbonate (PC), sodium glycocholate (SG), sodium lauryl sulphate (SLS), triacetin, triethyl citrate and tween 80 (TW 80) were added to the formulations either individually or in combination and studied for their effect on permeability and solution stability. Table 24 to 36 summarizes the formulations and permeation results of buffered and water formulations containing the above excipients.
TABLE 24
Results of the effects of Buffer and Water on Fentanyl Formulations.
INACTIVE
CONC. OF INGREDIENTS %
EXAMPLE FENTANYL BUFFER ALCOHOL PG Mannitol Triacetin TW80 PERMEATED
#* BASE OR WATER % (V) % (V) % (wt) % (wt) % (wt) IN 2 HOUR
12-a (b) 1 mg/ml B 20 5 17.33
(Control)
12-b (w) 1 mg/ml W 20 5 17.18
(Control)
12-c (b) 1 mg/ml B 20 5 0.1 0.15 16.14
12-d (w) 1 mg/ml W 20 5 0.1 0.15 12.10
12-e (b) 1 mg/ml B 20 5 0.3 0.2 16.02
12-f (w) 1 mg/ml W 20 5 0.3 0.2 12.35
12-g (b) 1 mg/ml B 20 5 0.15 0.3 0.1 16.43
12-h (w) 1 mg/ml W 20 5 0.15 0.3 0.1 9.76
12-i (b) 1 mg/ml B 20 5 0.2 0.2 18.74
12-j (w) W 20 5 0.2 0.2 13.19
12-k (b) 1 mg/ml B 20 5 0.3 14.73
12-l (w) 1 mg/ml W 20 5 0.3 13.76
(b)—buffer,
(w)—water
The results in Table 24 indicate that all the buffered formulations had similar permeability characteristics as that of control formulations except the buffered formulation containing 0.3% Tween 80 which showed lower permeability. All water formulations exhibited lower permeability than buffered formulations.
TABLE 25
Effect of HPBCD on Fentanyl Formulations
HPBCD
CONC. OF (MOLECULAR %
FENTANYL ALCOHOL PG RATIO TO PERMEATED
EXAMPLE #* BASE % (V) % (V) API) IN 2 HOURS
12-m (w) 1 mg/ml 20 5 17.18
(Control)
12-n (w) 1 mg/ml 20 5 1:1 (0.413%) 14.28
12-o (w) 1 mg/ml 20 5 1:2 (0.826%) 13.50
(b)—buffer,
(w)—water
TABLE 26
Effect of Mannitol on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL PG MANNITOL PERMEATED
EXAMPLE #* BASE % (V) % (V) % (WT) IN 2 HOURS
12-p (b) 1 mg/ml 20 5 17.33
(Control)
12-q (b) 1 mg/ml 20 5 0.3 17.71
12-r (b) 1 mg/ml 20 5 0.4 16.86
12-s (b) 1 mg/ml 20 5 0.5 15.41
12-t (b) 1 mg/ml 20 5 0.8 14.81
(b)—buffer,
(w)—water
TABLE 27
Effect of Polyvinyl Pyrrolidone (PVP) on Fentanyl Formulations
CONC. OF PG PVP %
FENTANYL ALCOHOL % % PERMEATED
EXAMPLE #* BASE % (V) (V) (WT) IN 2 HOURS
12-u (w) 1 mg/ml 20 5 17.18
(Control)
12-v (w) 1 mg/ml 20 5 0.1 16.73
12-w (w) 1 mg/ml 20 5 0.5 14.68
12-x (w) 1 mg/ml 20 5 1 14.52
12-y (w) 1 mg/ml 25 5 3 10.75
(b)—buffer,
(w)—water
TABLE 28
Effect of Propylene Carbonate (PC) on Fentanyl Formulations
CONC. OF PG PC %
FENTANYL ALCOHOL % % PERMEATED
EXAMPLE #* BASE % (V) (V) (WT) IN 2 HOURS
12-z (w) 1 mg/ml 20 5 17.18
(Control)
12-aa (w) 1 mg/ml 20 5 1 14.39
12-bb (w) 1 mg/ml 20 5 1.5 14.43
(b)—buffer,
(w)—water
TABLE 29
Effect of Sodium Glycocholate (SG) on Fentanyl Formulations
CONC. OF PG SG %
FENTANYL ALCOHOL % % PERMEATED
EXAMPLE #* BASE % (V) (V) (WT) IN 2 HOURS
12-cc (w) 1 mg/ml 20 5 17.18
(Control)
12-dd (w) 1 mg/ml 20 5 0.5 18.30
12-ee (w) 1 mg/ml 20 5 1 19.78
(b)—buffer,
(w)—water
TABLE 30
Effect of Triacetin on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL TRIACETIN PERMEATED
EXAMPLE #* BASE % (V) PG % (V) % (WT) IN 2 HOURS
12-ff (b) 1 mg/ml 20 5 17.33
(Control)
12-gg (b) 1 mg/ml 20 5 0.5 17.71
12-hh (b) 1 mg/ml 20 5 2 14.56
(b)—buffer,
(w)—water
TABLE 31
Effect of Triethyl Citrate on Fentanyl Formulations
CONC. OF TRIETHYL %
FENTANYL ALCOHOL CITRATE PERMEATED
EXAMPLE #* BASE % (V) PG % (V) % (WT) IN 2 HOURS
12-ii (w) 1 mg/ml 20 5 17.18
(Control)
12-jj (b) 1 mg/ml 20 5 0.5 15.81
12-kk (b) 1 mg/ml 20 5 2 10.02
(b)—buffer,
(w)—water
TABLE 32
Effect of Tween 80 (TW 80) on Fentanyl Formulations
CONC. OF ALCO- %
EXAMPLE FENTANYL HOL PG TW80 PERMEATED
#* BASE % (V) % (V) % (WT) IN 2 HOURS
12-ll (b) 1 mg/ml 20 5 17.33
(Control)
12-mm (b) 1 mg/ml 20 5 0.3 14.73
12-nn (b) 1 mg/ml 20 5 0.6 13.72
(b)—buffer,
(w)—water
TABLE 33
Effect of Labrasol & SLS on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL PG LABRASOL SLS PERMEATED
EXAMPLE #* BASE % (V) % (V) % (WT) % (WT) IN 2 HOURS
12-oo (w) 1 mg/ml 20 5 17.18
(Control)
12-pp (w) 1 mg/ml 20 5 6.5 8.25
12-qq (w) 1 mg/ml 20 5 3.0 0.5 9.31
12-rr (w) 1 mg/ml 20 5 6.5 0.5 8.63
(b)—buffer,
(w)—water
TABLE 34
Effect of Mannitol & Triacetin on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL PG MANNITOL TRIACETIN PERMEATED
EXAMPLE #* BASE % (V) % (V) % (WT) % (WT) IN 2 HOURS
12-ss (b) 1 mg/ml 20 5 17.33
(Control)
12-tt (b) 1 mg/ml 20 5 0.1 0.15 16.14
12-uu (b) 1 mg/ml 20 5 0.15 0.4 18.33
12-vv (b) 1 mg/ml 20 5 0.15 0.5 17.14
12-ww (b) 1 mg/ml 20 5 0.2 0.3 16.73
12-xx (b) 1 mg/ml 20 5 0.25 0.5 16.31
12-yy (b) 1 mg/ml 20 5 0.3 0.2 17.70
12-zz (b) 1 mg/ml 20 5 0.4 0.2 16.88
(b)—buffer,
(w)—water
TABLE 35
Effect of Mannitol & TW 80 on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL PG MANNITOL TW 80 PERMEATED
EXAMPLE #* BASE % (V) % (V) % (WT) % (WT) IN 2 HOURS
12-aaa (b) 1 mg/ml 20 5 17.33
(Control)
12-bbb (b) 1 mg/ml 20 5 0.2 0.2 18.74
12-ccc (b) 1 mg/ml 20 5 0.3 0.1 16.53
12-ddd (b) 1 mg/ml 20 5 0.3 0.2 16.02
(b)—buffer,
(w)—water
TABLE 36
Effect of Triacetin & TW 80 on Fentanyl Formulations
CONC. OF %
FENTANYL ALCOHOL PG TW 80 TRIACETIN PERMEATED
EXAMPLE #* BASE % (V) % (V) % (WT) % (WT) IN 2 HOURS
12-eee (b) 1 mg/ml 20 5 17.33
(Control)
12-fff (b) 1 mg/ml 20 5 0.1 0.2 16.99
12-ggg (b) 1 mg/ml 20 5 0.1 0.3 16.63
(b)—buffer,
(w)—water
The results indicate that addition of individual excipients including HPBCD (Table 25), PVP (Table 27), PC (Table 28), Triethyl citrate (Table 31) and Tween 80 (Table 32) to the formulation decreased the permeability of fentanyl across MatTek buccal membranes irrespective of excipient concentration. As shown in Table 26 and Table 30, formulations containing 0.3% Mannitol and 0.5% Triacetin showed similar permeability characterisitics as that of control formulation but the permeability decreased as the concentrations of these individual excipients were increased in the formulations. Stability studies indicated that a minimum of 0.45% and 0.5% mannitol concentration should be added to buffer and water formulations, respectively, to keep them stable. In case of Triacetin, formulations containing 0.5% or higher concentrations of triacetin were found to be stable.
Permeation of fentanyl from formulations containing SG was greater than that of control formulations (Table 29). Also, we observed that fentanyl permeation increased with the increase of SG concentration in the formulation.
The results of fentanyl permeation across MatTek buccal tissues from formulations containing combination of excipients are shown in Tables 33-36. Addition of Labrasol to the formulation improved the stability but decreased fentanyl permeation across MatTek buccal tissues. Similar results were observed with formulations containing labrasol and SLS. Among all the formulations containing combination of excipients, two formulations, Example 12-uu (0.15% mannitol, 0.4% triacetin) and Example 12-bbb (0.2% mannitol, 0.2% TW80), showed higher permeability compared to control formulation. We observed that the presence of mannitol in triacetin formulations did not show any improvement in the permeation. Hence, the formulation containing 0.5% triacetin (Table 30) was selected for further studies. Though the formulation, Example 12-bbb, showed good permeability mannitol concentration was increased to 0.3% to improve the stability of the product.
Both fentanyl citrate and fentanyl base formulations were stable at all temperatures studied. The data from in-vitro tissue permeation studies, as shown in Table 3, showed that permeation of fentanyl from fentanyl base formulations was about 10-fold higher than from fentanyl citrate formulations. Water and buffer formulations did not show any significant difference in fentanyl base permeation across buccal tissue. Our studies also showed that fentanyl base formulation containing Miglyol had very low permeability. Among the excipients, triacetin at 0.5% and mannitol at 0.3% in combination with 0.2% TW 80 showed good permeability and stability.
The transport of fentanyl across buccal tissues was dependent on ethyl alcohol (ethanol) and propylene glycol (PG) concentrations in the formulation. Ethanol is used as a cosolvent to solubilize fentanyl base in aqueous based formulations. In this investigation, we observed that formulations (with 1 mg/ml of fentanyl base) containing less than 20% of ethanol were precipitating at refrigerated conditions. Also, we observed that permeation of fentanyl across buccal tissue was indirectly proportional to ethanol concentrations in the formulations (Table 21). Similar results were observed with PG (Table 22). PG was helpful in increasing the solubility of fentanyl base in aqueous solution and also in enhancing the permeation of the fentanyl base across buccal tissues. Formulations containing more than 5% PG did not show significant improvement in the permeation of fentanyl, however, the solution viscosities increased proportionally with PG concentrations. The formulations containing more than 5% PG did not show good spray characteristics.
Example 13
In Example 13, a formulation having the following ingredients in Table 37 was tested. The formulation did not include fentanyl.
TABLE 37
Ingredient
Dehydrated Alcohol % (v/v)  30%
Propylene glycol % (v/v)   5%
Peppermint Oil % (v/v) 0.2%
Borate buffer (pH 9) QS
Hydroxy Propyl Beta 1:2
Cyclodextrin (HPBCD)
The formulation was sprayed using a 0.10 ml multidose nasal spray pump by Pfeiffer of America, Princeton, N.J. and the droplets were measured using a Malvern Mastersizer S device, by Malvern Instruments Ltd. A single depression of the sublingual spray pump generated a plume which was then analyzed for spray particles. The sample size for the dose volume, spray pattern, and droplet size distribution was 25 sprays.
Droplet Volume
In the droplet volume evaluation, 25 spray samples were evaluated using 5 different stroke numbers for each spray sample the following results were measured:
Overall mean=100.4 μl
Maximum single actual value=103.2 μl
Lowest single actual value=95.3 μl
Standard deviation=1.1
Range=7.9
Coefficient of variation=1.1%
Spray Pattern
In the spray pattern evaluation, 25 spray samples were evaluated using a manual actuation at 30 mm from the target. The formulation was dyed with methylene blue and the following spray pattern results were measured:
Small Diameter [mm]
min: 35.4
mean: 50.6
max: 62
s: δ 7.00
Largest Diameter [mm]
min: 40
mean: 56.9
max: 67
s: δ 6.01
spray angle
min: 64°
mean: 83.3°
max: 94°
s: 0.703
Ratio (Largest/Smallest Diameter)
min: 1.04
mean: 1.13
max: 1.33
s: 0.073
Droplet Size Distribution
In the droplet size distribution evaluation, 25 spray samples were evaluated using a manual actuation at 30 mm from the target. The following droplet size distribution results were measured:
Percentage Share of Droplet Diameters at 10 μm [%]
min 0.65
mean: 1.66
max: 2.70
s: 0.527
10% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 15
mean: 18.2
max: 23
s: 1.91
50% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 35
mean: 44.7
max: 65
s: δ 7.52
90% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 96
mean: 154.4
max: 349
s: 64.42
Example 14
In Example 14, a formulation having the following ingredients in Table 38 was tested. The formulation did not include fentanyl.
TABLE 38
Ingredient
Dehydrated Alcohol % (v/v) 30% 
Propylene glycol % (v/v) 5%
Peppermint Oil % (v/v) 0.2%  
Borate buffer (pH 9) QS
Hydroxy Propyl Beta Cyclodextrin 1:2
(HPBCD)
Hydroxy Propyl Cellulose (HPC EP) 1%
% (v/v)
The formulation was sprayed using a 0.10 ml multidose nasal spray pump by Pfeiffer of America, Princeton, N.J. and the droplets were measured using a Malvern Mastersizer S device, by Malvern Instruments Ltd. A single depression of the sublingual spray pump generated a plume which was then analyzed for spray particles. The sample size for the dose volume, spray pattern, and droplet size distribution was 25 sprays.
Droplet Volume
In the droplet volume evaluation, 25 spray samples were evaluated using 5 different stroke numbers the following results were measured:
Overall mean value=101.5 μl
Maximum single actual value=103.7 μl
Lowest single actual value=96.1 μl
Standard deviation=1.3
Range=7.6
Coefficient of variation=1.2%
Spray Pattern
In the spray pattern evaluation, 25 spray samples were evaluated using a manual actuation at 30 mm from the target. The formulation was dyed with methylene blue and the following spray pattern results were measured:
Small Diameter [mm]
min: 29
mean: 34.0
max: 46
s: δ 3.47
Largest Diameter
min: 35
mean: 40.8
max: 55
s: δ 4.13
Spray Angle
min: 580
mean: 63.80
max: 80°
s: 4.65
Ratio (Largest/Smallest Diameter)
min: 1.08
mean: 1.20
max: 1.50
s: 0.092
Droplet Size Distribution
In the droplet size distribution evaluation, 25 spray samples were evaluated using a manual actuation at 30 mm from the target. The following droplet size results were measured:
Percentage Share of Droplet Diameters at 10 μm [%]
min: 0.24
mean: 0.68
max: 1.28
s: δ 0.278
10% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 22
mean: 26.7
max: 35
s: δ 2.95
50% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 61
mean: 83.3
max: 114
s: 11.41
90% of the Droplet Diameters are Smaller than the Indicated Value [μm]
min: 192
mean: 294.6
max: 440
s: 53.32
Example 15
In Example 15, an assay procedure for fentanyl in 1, 2, 4, 6, and 8 mg/mL fentanyl sublingual spray samples with a working concentration of between 0.1 μg/mL and 5 μg/mL fentanyl in solution was performed. This method was developed and qualified in compliance with GMP requirements. The method was determined to be linear over the range of 0.05 μg/mL to 7.83 μg/mL fentanyl. The fentanyl working standard solution was stable over a seven day period in volumetric glassware and amber HPLC vials at refrigerated and ambient conditions.
The equipment and supplies utilized in this process included an HPLC system equipped with a pump, variable wavelength detector, and autosampler, or equivalent, a Waters Symmetry HPLC column (C18, 4.6×75 mm, 3.5 μm particle size), 0.45 μm, 47 mm nylon filters (Gelman Nylaflo® P/N 66608 or equivalent), acetonitrile (HPLC Grade), potassium phosphate monobasic (ACS Grade), phosphoric acid (ACS Grade), deionized water, alcohol (ethanol, absolute), and fentanyl base reference standard.
The solution preparations were prepared as described below and may be scaled as required.
For each liter of phosphate buffer solution (50 mM KH2PO4 pH 2.8), 6.8 g of potassium phosphate mono basic and 1 liter of water was combined in a suitable vessel and mixed well. The pH of the solution was adjusted to 2.8 with the drop-wise addition of phosphoric acid. The solution was filtered through 0.45 μm nylon. This solution expires after one month.
For each liter of mobile phase (25% ACN, 75% phosphate buffer) solution, 750 mL of phosphate buffer solution was combined with 250 mL of acetonitrile in a suitable vessel and mixed well. The system is degassed by an appropriate method before use if required. This solution expires after one month.
For each liter of stock diluent (95/5, Ethanol/Acetonitrile) solution, 950 mL ethanol and 50 mL acetonitrile was combined in a suitable container and mixed well. This solution expires after one month.
Stock standard I, 40 μg/mL, (“SSI”), was prepared by weighing approximately 10.0 mg of fentanyl reference standard and added to a 250 mL volumetric flask. Approximately 200 mL stock diluent was added and swirled to dissolve the solid material. The mixture was diluted to the desired volume with stock diluent and mixed well.
Stock standard II, 40 μg/mL, (“SSII”), was prepared by weighing approximately 10.0 mg of fentanyl reference standard and add to a 250 mL volumetric flask. Approximately 200 mL of stock diluent was added and swirled to dissolve solid material. The mixture was diluted to the desired volume with stock diluent and mixed well.
Working standard I, 2.4 μg/mL, (“WSI”), was prepared by transferring 3.0 mL of stock standard I to a 50 mL volumetric flask. The mixture was diluted to the desired volume with mobile phase solution and mixed well. This solution expires after seven days.
Working standard II, 2.4 μg/mL, (“WSII”) was prepared by transferring 3.0 mL of stock standard II to a 50 mL volumetric flask. The mixture was diluted to the desired volume with mobile phase solution and mixed well. This solution expires after seven days.
The chromatographic conditions for the HPLC procedure are set forth below:
  • Column: HPLC Column Waters Symmetry C18, 4.6×75 mm, 3.5 μm particle size
  • UV Detection: 214 nm
  • Flow Rate: 2.0 mL/minute
  • Injection Volume: 50 μL
  • Temperature: Ambient (The temperature may be controlled at 25° C.)
  • Acquisition Time: 13 minutes
    HPLC Procedure
After the system suitability is established, a maximum of 12 sample solutions can be injected in between working standards. A typical sequence would be as follows:
2x Mobile Phase (Blank)
2x Working standard II
5x Working standard I
Ix Sample (up to 12 injections)
Ix Working standard I
Ix Sample (up to 12 injections)
Ix Working standard I
System Suitability
There should be no significant interfering peaks present at the retention time of fentanyl in the mobile phase blank injections. In terms of injection precision, the RSD of fentanyl for five replicate injections of working standard I should not exceed 2.0%. In terms of standard agreement, the agreement between the average peak response for the first five working standard I injections and the two working standard II injections should be between 98 to 102%. The agreement between working standards I and II need only be demonstrated once during the expiry of standards. The tailing factor at 5% peak height for fentanyl in the first working standard I injection should be between 0.8 and 1.5. In terms of standard precision over the run, the RSD of peak area for fentanyl in the working standard I injections over the run (OTR) should not exceed 2.0%.
Calculations are performed as set forth below.
Working standard concentration is calculated as follows:
Mass of standard ( mg ) 250.0 mL * 1000 µg 1 mg * 3.0 mL 50.0 mL * Purity of Standard = µg / mL Fentanyl
Response Factor (Rf) is calculated as follows:
Fentanyl Peak Area Fentanyl Concentration ( µg / mL ) *= R f
Standard Agreement is calculated as follows:
Average R f WSII Average R f WSI * 100 = % Standard Agreement
Example 16
In Example 16 the method for determination of droplet size distribution by laser diffraction for fentanyl sublingual spray using the Spraytec device by Malvern was performed.
All data generated and described within this report were reviewed for compliance with Good Manufacturing Practices (21 CFR Parts 210 and 211).
The purpose of this project was to develop and validate a droplet size distribution method by laser diffraction for use with fentanyl sublingual spray product and placebo. The first portion of the project performed product evaluations to determine the proper automated actuation parameters to be used with the MightyRunt Actuation Station by Innova Systems, Inc. Using the automated actuation station, development of the droplet size distribution method for the sublingual product included vignetting studies, exhaust placement studies, and device placement studies.
The method validation evaluated intermediate precision between two analysts performing the developed method. All method development and qualification activities were performed using placebo.
Samples were prepared using Pfeiffer unit dose glass vials, Pfeiffer unit dose VI stoppers, Pfeiffer vial holder, and Pfeiffer unit dose applicator. The instrumentation utilized in the study include a Spraytec with 200 mm lens by Malvern Instruments, Inc, a MightyRunt Actuation Station by Innova Systems Inc. equipped with an exhaust fan attachment, and a Mettler Toledo balance Model AT201.
Actuation Parameter Study
Using the Spraytec to track the plume duration and droplet size distribution, the actuation parameters for the MightyRunt Actuation Station (MightyRunt) were optimized to replicate the plume duration, droplet size distribution, and shot weight generated by manual actuation. Dv10 (10% of the droplet diameters are smaller that the indicated value), Dv50 (50% of the droplet diameters are smaller that the indicated value), and Dv90, (90% of the droplet diameters are smaller that the indicated value), results from six devices with manual actuations were compared with the results from the six devices with automated actuations.
Acceptance Criteria
The individual shot weight results for the automated actuations should all fall within the range of 75%-125% of the average shot weight for the manual actuations. The average Dv10, Dv50 and Dv90 results of the automated actuations should be within 75% to 125% of the average Dv10, Dv50 and Dv90 results for the manual actuations.
Statistical analysis shall include performance of a students' t-test on the two sets of droplet size distribution results. The results of the students' t-test should indicate that the manual versus automated sets of data is statistically equivalent.
Method Development
Method development involved a vignetting study and exhaust study utilizing the Spraytec. Actuations were performed using the MightyRunt and previously determined parameters. Vignetting occurs during laser diffraction analysis when the small droplets of a spray plume scatter the laser at an angle too steep to be captured by the range lens. Placement of the device close enough to the range lens to capture all of the scattered light without deposition on the range lens is critical. The vignetting study determined the appropriate range lens for analysis and the appropriate distance from device to range lens.
Exhaust placement affects plume travel. Plume velocity should not be accelerated by the draw of the exhaust and large droplets should not fall while traveling through the laser path. The exhaust study determined the appropriate position behind the plume of the spray to ensure proper plume capture after passing through the laser path. The method included two distances for analysis from the tip of the nozzle to the path of the laser for a more complete characterization of the droplet size distribution.
Method Validation
Validation consisted of determining the precision and ruggedness of the method. A total of 24 devices from a single lot of placebo were used in the validation. One analyst tested 6 actuations at each distance. To demonstrate ruggedness, a second analyst repeated the analyses. Dv10, Dv50, and Dv90 results were compared.
Acceptance Criteria
The individual shot weight results for analyst one should all fall within the range of 75%-125% of the average shot weight for analyst two. The average Dv10, Dv50 and Dv90 one should be within 75% to 125% of the average Dv10, Dv50 and Dv90 results for analyst two.
The final experimental procedure is set forth below.
Prior to analysis, the background and scattering profiles were verified as appropriate for analysis.
The sample bottle is inserted into the nozzle holder. Coasters were used to raise the platform for minimal adjustment. The coaster attached to the device holder was placed on top of coasters used for adjustment.
The MightyRunt with bottle and exhaust fan was placed in the appropriate positions for analysis. The MightyRunt was raised with the nozzle centered in front of the laser path with a lab jack. It was ensured that the MightyRunt was level following adjustment. Two bottle-to-laser distances were be evaluated, 7 cm and 4 cm, measured from the pump tip to the center of the laser path. For all analyses, the bottle was placed 14 cm from the range lens support structure to the pump tip.
The exhaust fan was turned on and placed on a lab jack 3 cm behind the laser path, centered behind the device, measured from the center of the laser path to the front edge of the exhaust shield. The exhaust fan had an impaction surface for the droplets to adhere to, i.e. a cheesecloth placed in the path of the droplets. ⅛ sheet of 18×36 inch cheesecloth folded into approximately a 4.5-inch square (4 layers of cheesecloth) is a sufficient impaction surface and will not impede the flow of the fan.
The device was actuated using the MightyRunt station and droplet size distribution was collected for each individual shot at the appropriate distances for analysis.
A report was printed for the entire plume duration and the plume plateau.
Data Reporting
The data for the D10, D50, D90, and Span values for each actuation are reported. The average and precision (% RSD) (percent relative standard deviation) were calculated for the D10, D50, and D90. Note: Span is defined as (D90-D10)/D50. Individual droplet size distribution results (μm) and span values (unitless) were reported to X.XX. All average droplet size distribution results should be reported to X.X μm. All RSD values should be reported to X.X %.
Statistical analysis included performance of a students' t-test on the two sets of droplet size distribution results. The results of the students' t-test should have indicated that the analysts sets of data is statistically equivalent.
Results and Discussion
All spray plumes have three stages. The first stage, development, was characterized by variable droplets and decreasing transmission of the laser. The second stage, stable, was characterized by a stable droplet distribution and transmission. Variable droplets and increasing transmission characterize the final stage, dissipation. All comparisons of droplet size distribution used the stable stage of the plume.
Actuation Parameter Study
During the optimization of the actuation parameters for the MightyRunt, the plumes from manual and automated actuations were measured using the Spraytec. Dv10, Dv50, and Dv90 results were compared to optimize the automated parameters. The type of device that was examined in this study was significantly different from typical nasal sprays. Rather than a spring requiring a consistent force to initiate actuation and deliver the drug, there was an amount of force that was required to break the tabs or actuate the device. This force was not the same amount of force required to deliver the drug from the device once the tabs are broken. Because this device did not contain a spring, the parameters of interest were actuation force, force rise time, and minimum travel distance. The finalized MightyRunt parameters are listed below in Table 39 and the utilized Spraytec settings are listed below in Table 40.
TABLE 39
Mighty Runt Actuation Parameters
Parameter Setting
Actuation Force 4.0 kg
Force Rise Time 0.2 sec
Hold Time 1.0 sec
Force Fall Time 1.0 sec
Spray Delay
1 sec
Minimum Travel Distance 10 mm
Maximum travel Time 4.0 sec
Trigger Signal Delay 0.0 sec
Stage Yes
TABLE 40
Spraytec Settings
Option Setting
Test Duration
 200 msec
Data Acquisition Rate 1000 Hz
Acquisition Duty Cycle  0%
Experimental Trigger Transmission
Transmission Trigger 98%
Range Lens
 200 mm
The comparison of manual and automated actuations was performed using six devices with manual actuations and six devices with automated actuations. The droplet size distribution and shot weight results are summarized in Table 41 below.
TABLE 41
Shot
Weight %
Device D10 (μm) D50 (μm) D90 (μm) (mg) Average
Manual
1 19.0 44.5 76.8 89.1
Manual 2 18.1 42.7 84.9 86.9
Manual 3 33.3 58.7 108.4 90.4
Manual 4 20.8 44.6 81.2 87.0
Manual 5 18.3 42.7 80.7 87.8
Manual 6 19.0 44.3 81.5 84.9
Average 21.4 46.3 85.6 87.7
% RSD 27.5 13.3 13.4 2.2
Mighty Runt 1 17.3 37.5 65.1 83.8 96
Mighty Runt 2 16.1 35.3 64.0 66.0 75
Mighty Runt 3 18.8 38.9 66.2 84.2 96
Mighty Runt 4 15.4 37.8 77.6 85.4 97
Mighty Runt 5 17.6 38.6 67.1 82.4 94
Mighty Runt 6 19.4 41.9 74.8 88.0 100
Average 17.4 38.3 69.1 81.6
% RSD 8.8 5.6 8.2 9.7
Overall 19.43 42.30 77.36 84.7
average
Overall % RSD 23.7 14.3 15.7 7.5
Automated 81 83 81
Actuation
Average as %
of Manual
Actuation
Average
t-stat 1.60 2.97 3.15
t-critical 1.94 1.94 1.81
Result Same Different Different
population population population
Acceptance Criteria
Shot weight results for the automated actuations ranged from 75%-100% of the average for manual actuations, meeting the acceptance criteria of 75%-125% of the average shot weight for the manual actuations. The average Dv10 for automated actuations was 81% of the average Dv10 for manual actuations. The average Dv50 for automated actuations was 83% of the average Dv50 for manual actuations. The average Dv90 for automated actuations was 81% of the average Dv90 for manual actuations. Each of these parameters met acceptance criteria of 75%-125% of the average for manual actuations. Students' t-tests, while not necessarily appropriate for the small data sets, indicated that the data sets for manual actuations and automated actuations were equivalent for Dv10, but not for Dv50 or Dv90. It was not possible to accurately replicate the droplet size distribution from manual actuations with the MightyRunt. The force required to reliably break the tabs and actuate the devices produced a distribution with smaller droplets than that of the manual actuations. Less aggressive actuation parameters, which should produce larger droplet sizes, were not sufficient for consistent actuation of the devices. Spraytec method development proceeded with these parameters despite not meeting the acceptance criteria specified in the protocol.
Method Development
Method development involved a vignetting study, device placement study and exhaust placement study utilizing the Spraytec. Actuations were performed using the MightyRunt and the previously qualified parameters. For all tests, the device was placed in front of the laser beam path, the plume traveled through the laser path, and the plume was collected in an exhaust manifold placed behind the laser beam.
The vignetting experiments were performed with the device aligned with the front of the instrument (approximately 10 cm from the beam) and varying distances for the device-to-range lens placement. The results are summarized in Table 42.
TABLE 42
Distance to Range
Lens (cm) Dv10 (μm) Dv50 (μm) Dv90 (μm)
6 17.2 38.6 65.9
16.1 36.5 63.0
8 18.6 41.3 71.6
21.9 44.2 84.6
10 16.7 38.7 80.4
16.9 39.8 68.0
12 22.1 46.1 89.8
17.1 37.1 65.5
15.6 35.7 62.7
14 17.4 42.3 72.5
16.6 36.5 64.2
A plot of the Dv10, Dv50, Dv90, and plume records values versus placement is set forth in FIG. 11. The data showed no significant trend over the entire range of placements. To minimize the possibility of deposition of droplets on the range lens during testing, the placement of 14 cm (approximately centered between the laser and range lens supports) was chosen and used for all further testing.
To evaluate exhaust placement on the plume during testing, PSD (particle size diameter) data was collected and evaluated at four distances for exhaust-to-laser beam. The data are summarized in Table 43 below.
TABLE 43
Exhaust Height
(cm) Dv10 (μm) Dv50 (μm) Dv90 (μm)
7 cm Device-to-Laser Beam Placement
No exhaust 14.4 28.1 51.7
13.3 28.6 54.0
3 19.8 37.1 60.9
18.1 36.3 61.0
4 cm Device-to-Laser Beam Placement
No exhaust 17.8 35.0 57.9
18.7 36.3 60.5
1 14.6 29.4 51.0
12.6 28.0 56.6
3 15.1 30.0 52.0
13.6 27.6 49.0
5 14.3 32.8 79.8
13.4 28.7 51.7
7 11.2 25.5 55.7
15.9 30.0 50.8
A graphical summary of Dv10, Dv50, and Dv90 values versus placement is included in FIG. 12. There were no significant changes in the Dv10, Dv50, Dv90 or plume duration values for plumes collected without exhaust, or with exhaust 1, 3, 5, or 7 cm behind the beam. The exhaust-behind-laser beam placement was chosen to be 3 cm to reduce the chance of deposition outside the fan housing of sprays performed at a device-to-beam distance of 7 cm.
The method was to include two distances for analysis from the tip of the device to the path of the laser for a more complete characterization of the droplet size distribution. Four distances were evaluated, and results are included in Table 44 below.
TABLE 44
Distance (cm) Dv10 (μm) Dv50 (μm) Dv90 (μm)
7 16.7 34.5 67.8
15.8 33.4 60.0
6 17.8 35.0 57.9
18.7 36.3 60.5
5 19.1 35.5 58.5
16.9 32.4 55.1
4 11.8 27.8 57.5
13.0 28.1 50.3
3 14.4 28.1 51.7
13.3 28.6 54.0
A graphical summary of Dv10, Dv50, and Dv90 values versus placement are included in FIG. 3. There was no observable trend in the data from various heights. While there is no guidance document for sublingual sprays, the FDA Guidance Document “Bioequivalence and Bioavailability Studies for Nasal Aerosols and Nasal Sprays for Local Action”, specifies two distances from 3-7 cm. The larger pair of distances (4 cm and 7 cm) from the beam was chosen in order to characterize a more fully developed plume.
Method Validation
An analyst tested six devices at both the 4 cm and 7 cm distances. A second analyst repeated the analyses at both distances with the next actuations after priming from each device on a second day. Validation results are summarized in Tables 45 and 46 below where the Dv10, Dv50, and Dv90 results were compared.
TABLE 45
Validation PSD Results at 4 cm Device-to-Laser Beam Placement
Shot
Dv10 Weight %
Device (μm) Dv50 (μm) Dv90 (μm) (mg) Average
Analyst
1
1 18.9 31.2 52.3 60.4 76
2 16.4 30.8 52.6 78.8 100
3 17.2 29.8 51.3 65.0 82
4 18.3 31.6 54.2 80.1 101
5 13.5 28.5 51.7 81.6 103
6 15.0 30.1 52.3 77.2 98
Average 16.5 30.3 52.4 73.9
% RSD 12.3 3.7 1.9 12.0
Analyst 2
1 14.2 28.8 57.5 76.8
2 11.3 26.8 54.9 76.9
3 11.8 27.2 52.2 79.9
4 15.4 29.2 52.6 80.0
5 12.3 27.8 60.2 76.9
6 11.8 26.8 52.2 84.5
Average 12.8 27.8 54.9 79.2
% RSD 12.8 3.7 6.0 3.8
Overall 14.7 29.0 53.7 76.5
Average
Overall 17.9 5.8 5.0 9.0
% RSD
Analyst
1 129 109 95
Average as
% of
Analyst 2
t-stat 3.50 4.20 −1.80
t-critical 1.81 1.81 1.81
Result Different Different Same
Population Population Population
TABLE 46
Validation PSD Results at 7 cm Device-to-Laser Beam Placement
ShotWeight
File Dv10 (μm) Dv50 (μm) Dv90 (μm) (mg) % Average
Analyst 1-7 cm
a009 23.3 37.0 58.8 80.1 104
a010 16.1 34.0 57.3 76.9 99
a011 16.0 34.9 59.7 79.0 102
a012 17.6 34.0 57.1 80.7 104
a013 20.0 35.5 58.0 82.3 106
a014 19.6 36.1 57.2 80.6 104
Average 18.8 35.3 58.0 79.9
% RSD 14.8 3.4 1.8 2.3
Analyst 2-7 cm
b009 11.8 28.7 55.0 72.6
b010 17.0 33.9 61.7 79.8
b011 15.3 31.8 55.8 74.8
b012 13.8 31.1 54.6 79.7
b013 13.3 30.7 55.1 80.8
b015 16.6 32.8 56.7 76.3
Average 14.6 31.5 56.5 77.3
% RSD 13.8 5.8 4.7 4.2
Overall 16.7 33.4 57.2 78.6
Average
Overall % 18.9 7.3 3.6 3.6
RSD
Analyst
1 128 112 103
Average as %
of Analyst 2
Average
t-stat 2.93 4.25 1.32
t-critical 1.81 1.83 1.81
Result Different Different Same
Population Population Population
Acceptance Criteria
Analyst 1 shot weight results ranged from 76%-103% of analyst 2 average for the 4 cm distance, and 99%-106% for the 7 cm distance, meeting the acceptance criteria of 75%-125%. The average Dv10 for Analyst 1 was 129% of Analyst 2 for the 4 cm distance and 128% for the 7 cm distance. The average Dv50 for Analyst 1 was 109% of Analyst 2 for the 4 cm distance and 112% for the 7 cm distance. The average Dv90 Analyst 1 was 95% of Analyst 2 for the 4 cm distance and 103% for the 7 cm distance. While Dv10 did not meet acceptance criteria of 75%-125%, this is a measurement of the smallest droplets in the plume and a higher variability is not unexpected. Results for Dv50 and Dv90 met acceptance criteria of 75-125%. Students' t-tests, while not necessarily appropriate for the small data sets, indicated that the data sets for Analyst 1 and Analyst 2 were equivalent for Dv90 at both distances, but not for Dv10 or Dv50 at either distance.
Conclusion
A method for the droplet size distribution analysis by laser diffraction for use with fentanyl sublingual spray was developed and subsequently qualified. Acceptance criteria based on statistical analysis with students' t-test were deemed inappropriate for the small data sets. Method validation acceptance criteria for agreement of Analysts 1 and 2 were determined to be too narrow for the high variability associated with measurement of the smallest droplets in the plume (Dv10). While these criteria were not met, the method was deemed acceptable for use. The method is suitable for use within the operating parameters specified herein.
Example 17
In Example 17, a study was performed to determine the respirable dose less than 9 μm for fentanyl in 1, 2, 4, 6, and 8 mg/mL fentanyl sublingual spray samples with a working concentration of between 0.1 μg/mL and 5 μg/mL fentanyl in solution. The method used was qualified in compliance with GMP requirements. The sample solutions were determined to be stable over a seven-day period in volumetric glassware and amber HPLC vials at refrigerated and ambient conditions.
The HPLC process was consistent with the process described in Example 15 above. The materials and supplies utilized in the study included acetonitrile (HPLC Grade), potassium phosphate monobasic (ACS Grade), phosphoric acid (ACS Grade), deionized water, alcohol (ethanol, absolute), Short Stack Andersen Cascade Impactor set-up consisting of a 5-liter expansion chamber, induction port, stages 0, 1, 2, and after filter, a vacuum source, in-line flow meter (Sierra Top-Track or equivalent), VWR Sterile sampling bags, glass fiber filter, 8.1 cm, external calibrated flow meter (Dry-Cal Flow Meter or equivalent), and a pneumatic actuator (Innova Systems Mighty Runt or equivalent).
Solution Preparation
Solution preparations were prepared according to the methods described in Example 15 above and may be scaled as required.
The extraction solution was 50:50 (95/5, Ethanol/Acetonitrile:Water). For every liter of prepared solution, 475 mL ethanol, 25 mL acetonitrile and 500 mL of water was combined in a suitable container and mixed well. This solution expires after one month.
The phosphate buffer solution was prepared in a concentration of 50 mM KH2PO4 with a pH 2.8. For every liter of prepared solution, 6.8 g potassium phosphate mono basic and 1 liter of water is combined in a suitable vessel and mixed well. The pH of the solution was adjusted to pH 2.8 with the drop-wise addition of phosphoric acid. The solution was filtered through 0.45 μm nylon. This solution expires after one month.
The impactor set-up will consist of a 5-liter expansion chamber, induction port, stages 0, 1, 2, and filter prepared according to the following procedure.
The filter stage was placed onto the impactor base.
An 8.1 cm glass fiber filter was placed into the after filter stage and secured with a clean rubber o-ring.
A solid plate was placed on top of the filter stage and then stage 2 was placed in position.
A plate with center hole cutout was placed on top of stage 2 and then stage 1 was placed in position.
A plate with center hole cutout was placed on top of stage 1 and then stage 0 was in position.
The cone was placed in position and the impactor was secured with the hold down clamps.
The induction port was affixed to the cone and the 5 L expansion chamber was placed on top of the induction port.
Testing Set-Up
The testing instrumentation was set up by placing an in-line flow meter between the vacuum source and cascade impactor with appropriate tubing. A leak test was performed on the impactor. A flow through the impactor was started by opening the vacuum source and the flow was adjusted to approximately 28.3 L/min. A hand was placed over the spray actuation port on the expansion chamber. The flow rate as indicated on the in-line meter was expected to fall to zero. If a flow was still registered, the condition of the impactor a-rings was checked, and the test repeated. To set the flow rate, the expansion chamber was removed from the induction port and an external calibrated flow meter was attached to the induction port and the flow was started. The flow was adjusted to 28.3±1 L/min with the external calibrated meter and the measurement displayed on the in-line flow meter was recorded for using during the testing procedure.
Testing Procedure
Two devices were actuated for the 1 mg/mL product strength for each assay result. One device was actuated for the 2, 4, 6, and 8 mg/mL product strength. With the expansion chamber in place, the vacuum was started and the flow adjusted to the measurement obtained in during the testing set-up.
The pre-actuated weight of the device in grams to a minimum of 4 decimal places was recorded. The device was positioned so that the spray would travel directly toward the wall opposite the actuation port. The sublingual spray device was actuated into the expansion chamber with the automated pneumatic actuator method parameters listed in Table 47.
TABLE 47
Parameter Setting
Actuation Force (kg) 5.0
Force Rise Time (s) 0.1
Hold Time (s) 1.0
Force Fall Time (s) 1.0
Spray Delays (s) 1
Minimum Travel Distance (mm) 10.0
Maximum Travel Time (s) 4.0
Trigger Signal Delay (s) 0.0
Stage Yes
The post actuated weight of the device in grams is recorded to a minimum of 4 decimal places.
Extraction Procedure
Extraction was accomplished by breaking down the impactor set-up and extracting each component by the following procedures:
The plates and the filter were each extracted separately in bags with 10.0 mL of extraction solution. The sample was extracted by hand shaking and kneading for at least one minute. 6.0 mL of the extracted sample was transferred to a 10 mL volumetric flask and diluted to volume with phosphate buffer solution and mixed well. This is the sample solution. This solution expires after seven days.
The expansion chamber is inverted and place in a holder. The induction port and cone is inserted into the expansion chamber ground glass joint. Approximately 40 mL of extraction solution is rinsed through the cone and induction port into the expansion chamber. The solution is swirled in the expansion chamber in an effort to extract the entire interior surface. The solution is decanted into a 200 mL volumetric flask. The rinse is repeated two additional times for a total rinse volume of approximately 120 mL. The flask is brought to volume with phosphate buffer solution and mixed well. This is the sample solution. This solution expires after seven days.
Assay
The sample solutions are assayed as per the procedure set forth in Example 15 above. Calculations are performed as follows.
Response Factor ( Rf ) = Fentanyl Peak Area Fentanyl Concentration ( µg / mL )
The fentanyl in the plate and filter samples were determined according to the following calculation:
The amount of fentanyl in µg = A samp * D f WSI R f OTR A samp = Area of fentanyl in sample preparation D f = Dilution factor of fentanyl sample solution preparation ( 10.0 mL × 10.0 ml / 6.0 mL ) WSI R f OTR = Working standard I response factor over the run
The fentanyl in the cone, induction port and expansion chamber is determined according to the following calculation:
Amount of fentanyl in µg = A samp * A samp WSI R f OTR A samp = Area of fentanyl in sample preparation A samp = Volume of fentanyl in sample solution preparation ( 200 mL ) WSI R f OTR = Working standard I response factor over the run
The respirable dose for 2, 4, 6, and 8 mg/mL is calculated as follows:
Respirable dose in μg=Sum of the Drug Mass in Particle Size Fraction Less Than 9 μm (μg)
The respirable dose for 1 mg/mL is calculated as follows:
Respirable dose ( µg ) = Sum of the drug mass in particle size fraction less than 9 µg ( µg ) 2 ( number of actuations )
The respirable fraction is calculated as follows:
Percent ( % ) Respirable fraction = Drug mass in particle size fraction less than 9 µm ( µg ) ( respirable dose ) Total Drug Mass ( µg ) × 100
The particle size cutoff diameters for reporting is set forth in Table 48 below.
TABLE 48
Impactor Component Particle Size Grouping
Expansion Chamber,  ≧9 μm
Induction Port,
and Cone
Plate
0
Plate 1 9 μm > X ≧ 5.8 μm
Plate
2 <5.8 μm
F
The results for the fentanyl sample assay results in μg, the respirable dose in the particle size fraction less than 9 μm (as per Table 48) in μg, and the percent respirable dose less than 9 μm in percent to one decimal place were reported.
The Certificate of Analysis for the determination of the respirable 1 mg/mL dose set forth in Table 49 below.
TABLE 49
Determination of Respirable Dose, 1 mg/mL Fentanyl
Test
Assay of Fentanyl in
sublingual spray samples Specification
Determination of respirable Method Report Results
dose in fentanyl sublingual Described in Example 17 Report Results
spray by cascade impaction Average Total
Fentanyl Particle Shot Mass Respirable
CI (μg/ Size Groupings weight <9 μm dose <9 μm
Run Sample dose) groupings percent (mg) (μg) (μg)
1 Globe 76.5694 ≧9 μm 96.4 85.4 2.9 3.6
Plate 0 0.5479
Plate 1 0.6228 9 μm > X ≧ 0.8
5.8 μm
Plate
2 0.4746 <5.8 μm 2.9
Filter 1.8149
2 Globe 78.6941 ≧9 μm 96.6 84.0 2.8 3.4
Plate 0 0.6746
Plate 1 0.6217 9 μm > X ≧ 0.8
5.8 μm
Plate
2 0.5000 <5.8 μm 2.6
Filter 1.6740
3 Globe 78.0529 ≧ 9 μm 97.1 85.3 2.3 2.9
Plate 0 0.5082
Plate 1 0.5429 9 μm > X ≧ 0.7
5.8 μm
Plate
2 0.4185 <5.8 μm 2.2
Filter 1.3596
Average percent respirable dose 3.3
Example 18 Fentanyl SL Spray Formulations
In Example 18, formulations 18A-18E were prepared in accordance with the procedures set forth in the above examples. The formulations are set forth in Table 50 below.
TABLE 50
Quantity % w/w
Formulation Formulation Formulation Formulation Formulation
Component 18A 18B 18C 18D 18E
Fentanyl base 0.111 0.222 0.4444 0.6667 0.8889
(1 mg/mL) (2 mg/mL) (4 mg/mL) (6 mg/mL) (8 mg/mL)
Dehydrated 55.00 55.00 55.00 55.00 55.00
alcohol
Propylene glycol 5.00 5.00 5.00 5.00 5.00
L-Menthol 0.05 0.05 0.05 0.05 0.05
Xylitol 3.00 3.00 3.00 3.00 3.00
Purified water 36.839 36.728 36.5006 36.2833 36.0611
TOTAL 100 100 100 100 100
Example 19 Clinical Study
A Phase I, single-dose, open-label, randomized, three-period, three-treatment crossover study was conducted to compare the rate of absorption and bioavailability of Fentanyl Sublingual Spray 400 mcg to Actiq® 400 mcg and to Fentanyl Citrate Injection (IV) 100 mcg.
Subjects received each of the treatments below in randomized fashion during the three treatment periods, separated by a washout period of at least 7 days:
Treatment A: Fentanyl Sublingual Spray (Formulation 18C of Example 18)
    • Dose=1×400 mcg sublingual spray
    • Insys Therapeutics, Inc.
Treatment B: Actiq®
    • Dose=1×400 mcg oral transmucosal unit
    • Cephalon
Treatment C: Fentanyl Citrate Injection
    • Dose=1×100 mcg IV
    • Hospira, Inc
Blood samples (1×6 mL) were collected in vacutainer tubes containing EDTA as a preservative from the subjects at 0 (pre-dose) and at 5, 10, 20, 30, and 40 minutes after dosing and at 1.0, 1.25, 1.5, 2.0, 4.0, 6.0, 8.0, 10.0, 12.0, 16.0, 24.0, and 36.0 hours after dosing. The plasma samples were then analyzed for fentanyl using a validated liquid chromatography tandem mass spectrometry (LC-MS-MS) procedure. The method was validated for a range of 0.0250 to 5.00 ng/mL for fentanyl, based on the analysis of 0.500 mL of EDTA human plasma.
Data from 21 subjects who completed the study was subjected to pharmacokinetic and statistical analyses. Concentration-time data was transferred from Watson LIMS directly to WinNonlin Enterprise Edition (Version 4.0, Pharsight Corporation) using the Custom Query Builder option for analysis. Data was analyzed by noncompartmental methods in WinNonlin. Concentration-time data that were below the limit of quantification (BLQ) were treated as zero (0.00 ng/mL) in the data summarization and descriptive statistics.
In the pharmacokinetic analysis, BLQ concentrations were treated as zero from time-zero up to the time at which the first quantifiable concentration was observed; embedded and/or terminal BLQ concentrations were treated as “missing”.
Pharmacokinetic parameters were then calculated for each formulation using non-compartmental methods. Specifically, the following pharmacokinetic parameters were calculated: peak concentration in plasma (Cmax), time to peak concentration (Tmax), elimination rate constant (λz), terminal half-life (T1/2), area under the concentration-time curve from time-zero to the time of the last quantifiable concentration (AUClast), and area under the plasma concentration time curve from time-zero extrapolated to infinity (AUCinf).
The bioavailability (F) after oral/sublingual administration was also calculated. The bioavailability was calculated by using the following equation:
F = Dose IV * A U C extravascular Dose extravascular * A U C IV ,
where DoseIV and Doseextravascular are the IV and extravascular (sublingual spray and Actiq®) doses, respectively, and AUCIV and AUCextravascular are the areas under the plasma concentration-time profiles after IV and extravascular administration, respectively. Individual AUClast and AUCinf values after each treatment were used for the calculations.
To compare the fentanyl sublingual spray to Actiq®, analysis of variance (ANOVA) and the Schuirmann's two one-sided t-test procedures at the 5% significance level were applied to the log-transformed pharmacokinetic exposure parameters, Cmax, AUClast, and AUCinf. The 90% confidence intervals for the ratio of the geometric means (Test/Reference) were calculated.
The mean concentration-time data are shown in Table 51 and FIG. 15 below. Results of the pharmacokinetic and statistical analyses are shown in Table 52 and Table 53 below. The bioavailability estimates are summarized in Table 54 below.
TABLE 51
Fentanyl Concentration-Time Data after Administration of Fentanyl Sublinqual
Spray 400 mcg (Treatment A), Actiq ® 400 mcg (Treatment B) and Fentanyl Citrate
Injection 100 mcg (Treatment C)
Treatment A: Treatment B: Treatment C:
Fentanyl Sublinqual Spray Actiq ® Fentanyl Citrate Injection
400 mcg 400 mcg 100 mcg
Time Mean SD CV Mean SD CV Mean SD CV
(hr) n (ng/mL) (ng/mL) (%) n (ng/mL) (ng/mL) (%) n (ng/mL) (ng/mL) (%)
0.00 21 0.00 0.00 NC 21 0.00 0.00 NC 21 0.00 0.00 NC
0.08 21 0.168 0.189 112.68 21 0.00146 0.00669 458.26 21 0.749 0.607 81.08
0.17 21 0.440 0.300 68.30 21 0.0396 0.0511 129.24 21 0.730 0.240 32.84
0.33 21 0.513 0.242 47.24 21 0.220 0.150 68.04 21 0.494 0.185 37.44
0.50 21 0.606 0.288 47.45 21 0.324 0.112 34.56 21 0.416 0.137 33.01
0.67 21 0.613 0.238 38.81 21 0.360 0.127 35.10 21 0.376 0.108 28.64
1.00 21 0.670 0.264 39.32 21 0.444 0.170 38.26 21 0.327 0.0773 23.61
1.25 21 0.679 0.204 30.11 21 0.478 0.163 34.07 21 0.299 0.0689 23.04
1.50 21 0.694 0.203 29.19 21 0.533 0.198 37.23 21 0.275 0.0607 22.05
2.00 21 0.674 0.182 26.98 21 0.570 0.173 30.40 21 0.229 0.0475 20.74
4.00 21 0.449 0.126 28.05 21 0.363 0.146 40.04 21 0.141 0.0339 24.00
6.00 21 0.218 0.0793 36.31 21 0.185 0.0713 38.57 21 0.0693 0.0167 24.16
8.00 21 0.170 0.0673 39.72 21 0.132 0.0519 39.43 21 0.0502 0.0157 31.19
10.00 21 0.159 0.0720 45.44 21 0.123 0.0609 49.39 21 0.0489 0.0209 42.68
12.00 21 0.0886 0.0396 44.65 21 0.0710 0.0399 56.20 21 0.0188 0.0193 102.75
16.00 21 0.0614 0.0322 52.50 21 0.0497 0.0299 60.15 21 0.00877 0.0145 165.79
24.00 21 0.0484 0.0357 73.79 21 0.0345 0.0319 92.44 21 0.00777 0.0171 220.03
36.00 21 0.00876 0.0164 187.39 21 0.00465 0.0123 263.87 21 0.00 0.00 NC
Note:
Plasma samples analyzed using a bioanalytical method with a validated range 0.0250 to 5.00 ng/mL; concentrations below limit of quantification set to zero (0.00 ng/mL) in the data summarization
NC = Not calculated
TABLE 52
Pharmacokinetic Parameters of Fentanyl
Treatment A: Treatment B: Treatment C:
Fentanyl Sublinqual Spray Actiq ® Fentanyl Citrate Injection
400 mcg 400 mcg 100 mcg
Parameter n Mean SD CV % n Mean SD CV % n Mean SD CV %
Tmax (hr) 21 1.28 0.60 47.18 21 1.70 0.42 25.04 21 0.16 0.08 50.52
C max 21 0.813 0.252 31.01 21 0.607 0.185 30.55 21 0.929 0.515 55.48
(ng/mL)
AUC last 21 4.863 1.708 35.12 21 3.677 1.440 39.17 21 1.688 0.4114 24.38
(hr * ng/mL)
AUC inf 16 5.761 1.916 33.26 18 4.182 1.670 39.93 16 1.758 0.3822 21.74
(hr * ng/mL)
AUC Extrap 16 10.26 5.66 55.19 18 10.64 5.68 53.38 16 11.14 3.44 30.92
(%)
λz (hr−1) 16 0.0904 0.0571 63.16 18 0.1097 0.0532 48.51 16 0.1775 0.0662 37.30
T1/2 (hr) 16 9.98 4.41 44.14 18 7.89 3.72 47.15 16 4.50 1.94 43.02
Tlast (hr) 21 25.15 7.17 28.50 21 22.86 6.83 29.87 21 13.81 5.51 39.90
C last 21 0.0408 0.0117 28.74 21 0.0363 0.00953 26.23 21 0.0352 0.0117 33.08
(ng/mL)
TABLE 53
Statistical Analysis of the Log-Transformed Systemic Exposure Parameters of
Fentanyl Comparing Fentanyl Sublingual Spray 400 mcg (Treatment A) to Actiq ®
400 mcg (Treatment B)
Dependent Geometric Meana Ratio (%)b 90% CIc ANOVA
Variable Test Ref (Test/Ref) Lower Upper Power CV %
In (Cmax) 0.7865 0.5884 133.67 119.67 149.31 0.9527 20.85
In (AUClast) 4.6392 3.4767 133.44 121.47 146.58 0.9859 17.65
In (AUCinf) 5.5080 4.0420 136.27 121.21 153.20 0.9341 17.06
aGeometric Mean for Treatment A (Test) and Treatment B (Ref) based on Least Squares Mean of log-transformed parameter values
bRatio(%) = Geometric Mean (Test)/Geometric Mean (Ref)
c90% Confidence Interval
TABLE 54
Bioavailability of Fentanyl after Administration of Fentanyl Sublingual
Spray (Treatment A) and Actiq ® (Treatment B)
F (AUClast) F (AUCinf)
CV CV
Treatment n Mean SD (%) n Mean SD (%)
A 21 0.721 0.199 27.59 11 0.756 0.212 28.02
B 21 0.540 0.135 25.08 13 0.511 0.0973 19.05
It was concluded that Fentanyl SL intermediates between Actiq® and Fentanyl IV, as to time to reach threshold concentration, and that Fentanyl SL concentrations are about 40% higher than Actiq® (Bioavailability is 71+16% vs. 52±13% for Actiq®). Terminal half-life is similar in all three treatments (Fentanyl IV has an artifactually different decline beyond 10 hr because concentrations on many subjects fell below assay sensitivity (Assay sensitivity=0.050 ng/mL (50 pg/mL)).
It was further concluded, based on reports of time to onset for Actiq® (30 min) and Fentora (15 min), that threshold concentration of fentanyl is probably ˜0.3-4 ng/mL. Fentanyl SL reaches 0.3-0.4 ng/mL 10 minutes post dose, which is 20 minutes sooner than Actiq®.
Example 20 Dose Escalation Clinical Study
A dose escalation Phase I study on Fentanyl SL has been initiated. Formulation 18A, Formulation 18B, Formulation 18C, Formulation 18D, and Formulation 18E, all of Example 18 were used. Doses of 100 mcg, 200 mcg, 400 mcg, 600 mcg, and 800 mcg were administered to 40, 39, 42, 40, and 42 patients, respectively. The bio-analytical method was performed according to the procedure set forth in Example 19 above. The preliminary pharmacokinetic data is provided in Table 54 below, and is graphically represented in FIG. 16, FIG. 17 and FIG. 18.
TABLE 54
Dose Adj'ed Dose Adj'ed
N Cmax AUC (0-t) Cmax AUC
# ng/ml Tmax h ng · h/mL (to 100 mg) (to 100 mg)
100 mcg 40 0.193 1.12 0.919 0.193 0.919
200 mcg 39 0.379 1.04 1.94 0.189 0.971
400 mcg 42 0.796 0.970 4.58 0.199 1.15
600 mcg 40 1.15 0.987 6.65 0.192 1.11
800 mcg 42 1.56 1.06 8.93 0.195 1.12
The preliminary data appears to support the conclusion that Fentanyl SL has faster onset (60% Tmax in 10 minutes) and therefore pain relief than analgesic products currently available commercially (i.e., Actiq®, Fentora®, Rapinyl®, BEMA Fentanyl). The preliminary data also indicates that Fentanyl SL stays close to Tmax for 100 minutes translating to pain relief for a longer time.
Example 21 Pharmacokinetic Profile of Fentanyl Sublingual (SL) Spray
In Example 21, a five-treatment, five-sequence, five-period crossover study of fentanyl SL spray was conducted under fasted conditions in up to 70 healthy subjects. The objectives were to determine the pharmacokinetics of five difference doses (Part A), and to assess the impact of temperature and pH in the oral cavity on the relative bioavailability at a fixed dose (Part B). Healthy subjects had to meet pre-specified eligibility criteria. Plasma samples were obtained at time points of 0, 5, 10, 20, 30, 40 min, 1, 1.25, 1.5, 2, 4, 6, 8, 10, 12, 16, 24 and 36 h post-dose and analyzed for fentanyl using a validated LC-MS-MS procedure.
53 subjects were enrolled in part A. Administration of fentanyl SL spray was dose-proportional over the 100 mcg to 800 mcg dose ranges. Fentanyl concentrations increase rapidly following administration, being above the LLOQ within 5 minutes, reaching 60.6% of the peak plateau by 10 minutes and 86.6% of the peak plateau by 20 minutes post dose. Fentanyl concentrations showed a relatively long plateau about the peak value (>80% of Cmax) that lasted approximately 2 hours.
14 subjects were enrolled in part B. Varying the pH and temp of the oral cavity did not affect the PK profile. No SAEs were noted. AEs were observed in 31 subjects in part A. 46 were probably related to study treatment, and 29 were possibly related. During part B, AEs were observed in 9 subjects. 7 were probably related to study treatment, and 17 were possibly related. AEs were emesis or nausea.
The results of this study support the rationale for assessing efficacy in patients with breakthrough pain. The dose proportionality supports a rationale for predictable dosing favorable for titration.
Example 22 Comparative Bioavailability of Fentanyl Sublingual (SL) Spray IV Fentanyl Citrate and Actiq
In Example 22, a single-dose, open-label, randomized, three-period, three-treatment crossover study with a washout period of at least seven days between study periods was conducted at a phase I contract clinic under good clinical practice guidelines. 40 healthy volunteers were enrolled, having met pre-specified eligibility criteria. Subjects received a single dose of fentanyl SL spray 400 mcg, Actiq 400 mcg lozenge, and fentanyl citrate 100 mcg by IV injection over 5 minutes in 3 separate treatment periods. Plasma samples were obtained at time points of 0, 5, 10, 20, 30, 40 min, 1, 1.25, 1.5, 2, 4, 6, 8, 10, 12, 16, 24 and 36 h post-dose and analyzed for fentanyl using a validated LC-MS-MS procedure.
The results showed that compared to intravenous administration, the median value for absolute bioavailability of fentanyl SL spray was 60.8%; bioavailability of Actiq was 46.6%. The median value for relative bioavailability of Fentanyl SL Spray to that of Actiq was 135%. Systemic absorption of fentanyl SL was more rapid than Actiq. Subjects were monitored for any adverse events. AEs were reported in 15 of the 40 subjects. All of the AEs were mild. Two of the AEs were probably related to the study drug (both were sublingual burning at 400 mcg). Three of the AEs were possibly related to the study treatment (headache, dizziness, and dry throat all reported at 400 mcg).
The results of this study support the rationale for assessing efficacy in patients with breakthrough pain.
Many other variations of the present invention will be apparent to those skilled in the art and are meant to be within the scope of the claims appended hereto, including but not limited to the particular unit dose or bi-dose devices and the particle size range of fentanyl produced, as well as other numerical parameters described in the examples, and any combination thereof.

Claims (20)

What is claimed is:
1. A method of treating pain comprising administering to a patient in need thereof a propellant-free sublingual spray formulation comprising an effective amount of fentanyl, the formulation providing a mean Tmax of about 1.28+/−0.60; and wherein said sublingual spray formulation comprises droplets having a mean diameter of at least about 10 microns; wherein said sublingual spray formulations comprises:
from about 0.1% to about 0.8% by weight of fentanyl or a pharmaceutically acceptable salt thereof;
from about 50% to about 60% by weight of ethanol; and
from about 4% to about 6% by weight of propylene glycol.
2. The method of claim 1, wherein the concentration of fentanyl in the sublingual spray formulation is from about 1 mg/mL to about 8 mg/mL.
3. The method of claim 1, wherein the concentration of fentanyl in the sublingual spray formulation is selected from the group consisting of about 1 mg/mL, about 2 mg/mL, about 4 mg/mL, about 6 mg/mL and about 8 mg/mL.
4. The method of claim 1, which provides a mean Cmax of about 0.813 ng/ml+/−0.252 based on a sublingual dose of about 400 mcg fentanyl when administered to humans.
5. The method of claim 1, which provides a dosage amount of fentanyl selected from the group consisting of about 100 mcg, about 200 mcg, about 600 mcg and about 800 mcg, and provides a mean Cmax which is substantially dose proportional to the sublingual spray formulation containing 400 mcg fentanyl dosage amount, when administered to humans.
6. The method of claim 1, provides a substantially dose proportional mean Cmax based on a mean Cmax of about 0.813 ng/ml+/−0.252 for a 400 mcg fentanyl dose when administered to humans.
7. The method of claim 1, which provides a mean Tmax when administered to humans selected from the group consisting of: about 1.12 hours when the formulation provides a 100 mcg dose, about 1.04 hours when the formulation provides a 200 mcg dose, about 0.97 hours when the formulation provides a 400 mcg dose, about 0.987 hours when the formulation provides a 600 mcg dose, and about 1.06 hours when the formulation provides a 800 mcg dose.
8. The method of claim 1, which provides a plasma concentration after administration to humans selected from the group consisting of: about 60% of the mean Cmax in about 10 minutes, about 86% of the mean Ciax by about 20 minutes and a combination thereof.
9. The method of claim 1, which provides a plasma concentration after administration to humans that is greater than about 80% of the mean Cmax for about 2 hours.
10. The method of claim 1, wherein the sublingual spray formulation comprises 400 mcg of fentanyl, and provides one or more mean pharmacokinetic values selected from the group consisting of: AUClast 4.863+/−1.70821 hr*ng/mL, AUCinf 5.761+/−1.916 hr*ng/mL, and AUCextrap 10.26+/−5.66%, when administered to humans.
11. The method of claim 1, which provides a dosage amount of fentanyl when administered to humans which is substantially dose proportional to the dosage which contains about 400 mcg fentanyl selected from the group consisting of about 100 mcg, about 200 mcg, about 600 mcg, about 800 mcg, and provides one or more pharmacokinetic values selected from the group consisting of: mean AUClast, mean AUCinf, and mean AUCextrap.
12. The method of claim 1, which provides a substantially dose proportional mean AUClast based on a mean AUClast of about 4.863+/−1.70821 hr*ng/mL for a 400 mcg fentanyl dose when administered to humans.
13. The method of claim 1, wherein the sublingual spray formulation comprises a 400 mcg dose of fentanyl, providing a geometric mean ln(Cmax) of about 0.7865 ng/ml when a dose is administered to humans.
14. The method of claim 1, wherein the sublingual spray formulation comprises a 400 mcg dose of fentanyl, providing a mean F(AUClast) of about 0.721+/−0.199 ng/mL when a dose is administered to humans.
15. The method of claim 1, wherein the sublingual spray formulation comprises a 400 meg dose of fentanyl when a dose is administered to humans, providing a mean F (bioavailability) selected from the group consisting of: about 71%+/−16%, 0.721+/−0.199 based on AUClast and about 0.756+/−0.212 based on AUCinf, or combinations thereof.
16. The method of claim 1, wherein the sublingual spray formulation further comprises water.
17. The method of claim 1 wherein the sublingual formulation provides a substantially dose proportional mean Cmax based on a mean Cmax of about 0.813 ng/ml+/−0.252 for a 400 mcg fentanyl dose when administered to humans.
18. The method of claim 1 wherein the sublingual formulation provides a substantially dose proportional mean AUClast based on a mean AUClast of about 4.863+/−1.70821 hr*ng/mL for a 400 mcg fentanyl dose when administered to humans.
19. A method of manufacturing a sublingual spray formulation comprising an effective amount of fentanyl and at least one pharmaceutically acceptable excipient comprising:
admixing fentanyl, purified water and dehydrated alcohol and placing the mixture into a sublingual delivery device; the formulation providing a mean Tmax of about 1.28+/−0.60 hours when a dose is administered sublingually to humans and upon delivery, providing a mean aerodynamic particle size of at least about 10 microns; wherein said sublingual spray formulation comprises:
from about 0.1% to about 0.8% by weight of fentanyl or a pharmaceutically acceptable salt thereof;
from about 50% to about 60% by weight of ethanol; and
from about 4% to about 6% by weight of propylene glycol.
20. A method of treating pain comprising administering to a patient in need thereof a propellant-free sublingual spray formulation comprising an effective amount of fentanyl, the formulation providing a mean time to 60% maximum plasma concentration (Tmax) of fentanyl of about 10 minutes; and wherein said sublingual spray formulation comprises droplets having a mean diameter of at least about 10 microns; wherein said sublingual spray formulations comprises:
from about 0.1% to about 0.8% by weight of fentanyl or a pharmaceutically acceptable salt thereof;
from about 50% to about 60% by weight of ethanol; and
from about 4% to about 6% by weight of propylene glycol.
US12/221,333 2007-08-02 2008-08-01 Sublingual fentanyl spray Active 2030-06-06 US8486973B2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US12/221,333 US8486973B2 (en) 2007-08-02 2008-08-01 Sublingual fentanyl spray
US13/895,111 US8835459B2 (en) 2007-08-02 2013-05-15 Sublingual fentanyl spray
US14/448,752 US9241935B2 (en) 2007-08-02 2014-07-31 Sublingual fentanyl spray
US14/966,301 US9642844B2 (en) 2007-08-02 2015-12-11 Sublingual fentanyl spray
US15/478,499 US10610523B2 (en) 2007-08-02 2017-04-04 Sublingual fentanyl spray

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US96307607P 2007-08-02 2007-08-02
US96325307P 2007-08-03 2007-08-03
US12/221,333 US8486973B2 (en) 2007-08-02 2008-08-01 Sublingual fentanyl spray

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/895,111 Continuation US8835459B2 (en) 2007-08-02 2013-05-15 Sublingual fentanyl spray

Publications (2)

Publication Number Publication Date
US20090176834A1 US20090176834A1 (en) 2009-07-09
US8486973B2 true US8486973B2 (en) 2013-07-16

Family

ID=40305148

Family Applications (5)

Application Number Title Priority Date Filing Date
US12/221,333 Active 2030-06-06 US8486973B2 (en) 2007-08-02 2008-08-01 Sublingual fentanyl spray
US13/895,111 Expired - Fee Related US8835459B2 (en) 2007-08-02 2013-05-15 Sublingual fentanyl spray
US14/448,752 Expired - Fee Related US9241935B2 (en) 2007-08-02 2014-07-31 Sublingual fentanyl spray
US14/966,301 Active US9642844B2 (en) 2007-08-02 2015-12-11 Sublingual fentanyl spray
US15/478,499 Active 2029-01-21 US10610523B2 (en) 2007-08-02 2017-04-04 Sublingual fentanyl spray

Family Applications After (4)

Application Number Title Priority Date Filing Date
US13/895,111 Expired - Fee Related US8835459B2 (en) 2007-08-02 2013-05-15 Sublingual fentanyl spray
US14/448,752 Expired - Fee Related US9241935B2 (en) 2007-08-02 2014-07-31 Sublingual fentanyl spray
US14/966,301 Active US9642844B2 (en) 2007-08-02 2015-12-11 Sublingual fentanyl spray
US15/478,499 Active 2029-01-21 US10610523B2 (en) 2007-08-02 2017-04-04 Sublingual fentanyl spray

Country Status (10)

Country Link
US (5) US8486973B2 (en)
EP (1) EP2180844B1 (en)
AU (1) AU2008282743B2 (en)
CA (1) CA2698749C (en)
DK (1) DK2180844T3 (en)
ES (1) ES2668366T3 (en)
NO (1) NO2180844T3 (en)
PL (1) PL2180844T3 (en)
PT (1) PT2180844T (en)
WO (1) WO2009017837A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8835460B2 (en) 2006-01-25 2014-09-16 Insys Therapeutics, Inc. Sublingual fentanyl spray and methods of treating pain
US10265309B2 (en) * 2016-05-27 2019-04-23 Insys Development Company, Inc. Sublingual opioid formulations containing naloxone

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1642578B2 (en) 2000-07-31 2021-11-03 Takeda Pharma A/S Fentanyl salt composition for nasal administration
CN101111281B (en) 2005-02-01 2013-02-06 因特利杰克特有限公司 Devices, systems, and methods for medicament delivery
US8361026B2 (en) 2005-02-01 2013-01-29 Intelliject, Inc. Apparatus and methods for self-administration of vaccines and other medicaments
US8173666B2 (en) 2007-03-12 2012-05-08 Nektar Therapeutics Oligomer-opioid agonist conjugates
US10512644B2 (en) 2007-03-12 2019-12-24 Inheris Pharmaceuticals, Inc. Oligomer-opioid agonist conjugates
GB2477545B (en) * 2010-02-05 2011-12-21 Special Products Ltd A liquid morphine composition for buccal administration
GB2481619B (en) * 2010-06-30 2012-06-20 Londonpharma Ltd Formulations and delivery devices for the sublingual administration of opioids
US8939943B2 (en) 2011-01-26 2015-01-27 Kaleo, Inc. Medicament delivery device for administration of opioid antagonists including formulations for naloxone
US8627816B2 (en) 2011-02-28 2014-01-14 Intelliject, Inc. Medicament delivery device for administration of opioid antagonists including formulations for naloxone
CA2923811C (en) 2013-09-10 2021-07-27 Insys Pharma, Inc. Sublingual buprenorphine spray
US9867818B2 (en) 2013-09-10 2018-01-16 Insys Development Company, Inc. Sublingual buprenorphine spray
US9918981B2 (en) 2013-09-10 2018-03-20 Insys Development Company, Inc. Liquid buprenorphine formulations
US9839611B2 (en) 2013-09-10 2017-12-12 Insys Development Company, Inc. Sublingual buprenorphine spray
WO2015070049A1 (en) * 2013-11-08 2015-05-14 Insys Pharma, Inc. Sildenafil sublingual spray formulation
KR20220054908A (en) 2014-04-04 2022-05-03 인트라-셀룰라 써래피스, 인코퍼레이티드. Organic compounds
US9855234B2 (en) * 2014-07-08 2018-01-02 Insys Development Company, Inc. Diclofenac sublingual spray
US9517307B2 (en) 2014-07-18 2016-12-13 Kaleo, Inc. Devices and methods for delivering opioid antagonists including formulations for naloxone
CA2981615A1 (en) * 2015-04-03 2016-10-06 Insys Development Company, Inc. Sildenafil sublingual spray formulations
PT3407888T (en) 2016-01-26 2021-03-19 Intra Cellular Therapies Inc Organic compounds
PL3407889T3 (en) 2016-03-25 2021-11-22 Intra-Cellular Therapies, Inc. Organic compounds and their use in treating or preventing central nervous system disorders
US10906906B2 (en) 2016-12-29 2021-02-02 Intra-Cellular Therapies, Inc. Organic compounds
WO2018126140A1 (en) 2016-12-29 2018-07-05 Intra-Cellular Therapies, Inc. Organic compounds
JP7132939B2 (en) 2017-03-24 2022-09-07 イントラ-セルラー・セラピーズ・インコーポレイテッド Novel compositions and methods
MX2021002321A (en) 2018-08-31 2021-04-28 Intra Cellular Therapies Inc Novel methods.

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4244478A (en) 1979-06-27 1981-01-13 Mpl, Inc. Closure assembly for unit dose vial
WO1990007333A1 (en) 1989-01-06 1990-07-12 Riker Laboratories, Inc. Fentanyl containing aerosol compositions
US5219083A (en) 1992-04-02 1993-06-15 Sterling Winthrop Inc. Stopper for reduction of particulate matter
US5958379A (en) 1994-09-30 1999-09-28 Mika Pharma Gesellschaft Fuer Die Entwicklung Und Vermarktung Pharmazeutischer Producte Mbh Pharmaceutical composition
US5976504A (en) 1993-08-10 1999-11-02 The Boots Company, Plc Topical aerosol formulation for cooling of mammalian tissues
WO2000047203A1 (en) 1999-02-12 2000-08-17 Mqs, Inc. Formulation and system for intra-oral delivery of pharmaceutical agents
WO2001097780A2 (en) 2000-06-22 2001-12-27 Pharmasol Ltd Pharmaceutical compositions comprising an opioid analgesic
US20020160991A1 (en) 2000-12-04 2002-10-31 Liming Shao Orally-bioavailable formulations of fentanyl and congeners thereof
US20030039680A1 (en) 1997-10-01 2003-02-27 Flemington Pharmaceutical Corporation Buccal, polar and non-polar spray or capsule
US20030077228A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating endocrine disorders
US20030077229A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing cardiovascular or renal drugs
US20030082107A1 (en) 1997-10-01 2003-05-01 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating an infectious disease or cancer
US20030095926A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the gastrointestinal tract or urinary tract
US20030095925A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating metabolic disorders
US20030095927A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating muscular and skeletal disorders
US20030190286A1 (en) 1997-10-01 2003-10-09 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating allergies or asthma
WO2004016243A2 (en) 2002-08-16 2004-02-26 Generex Pharmaceuticals Inc. Methods of administering and enhancing absorption of pharmaceutical agents
US20040092428A1 (en) 2001-11-27 2004-05-13 Hongming Chen Oral pharmaceuticals formulation comprising paclitaxel, derivatives and methods of administration thereof
US20040120895A1 (en) 1997-10-01 2004-06-24 Novadel Pharma, Inc. Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the central nervous system
US6759059B1 (en) 1998-09-24 2004-07-06 Diabact Ab Fentanyl composition for the treatment of acute pain
US20040136913A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing sumatriptan
US20040136914A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing ondansetron
US20040136915A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing atropine
US20040141923A1 (en) 1997-10-01 2004-07-22 Dugger Harry A. Buccal, polar and non-polar spray containing alprazolam
WO2004075877A1 (en) 2003-02-24 2004-09-10 Pharmaceutical Productions, Inc. Transmucosal drug delivery system
WO2004080382A2 (en) 2003-03-11 2004-09-23 Arakis Ltd. Novel compositions containing fentanyl
US20040265239A1 (en) 1997-10-01 2004-12-30 Novadel Pharma Inc. Buccal, polar and non-polar spray containing zolpidem
US20050002867A1 (en) 1997-10-01 2005-01-06 Novadel Pharma Inc. Buccal, polar and non-polar sprays containing propofol
US20050163719A1 (en) 1997-10-01 2005-07-28 Dugger Harry A.Iii Buccal, polar and non-polar spray containing diazepam
US20050180923A1 (en) 1997-10-01 2005-08-18 Dugger Harry A.Iii Buccal, polar and non-polar spray containing testosterone
US6946150B2 (en) * 2002-08-14 2005-09-20 Gw Pharma Limited Pharmaceutical formulation
US20050281752A1 (en) 1997-10-01 2005-12-22 Dugger Harry A Iii Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the central nervous system
US20050287075A1 (en) 1997-10-01 2005-12-29 Dugger Harry A Iii Buccal, polar and non-polar spray or capsule containing drugs for treating pain
US20060062812A1 (en) 2003-03-11 2006-03-23 Calvin Ross Novel compositions
WO2007007059A1 (en) 2005-07-08 2007-01-18 Sosei R & D Ltd. Fentanyl formulation containing an essential oil
US20070261695A1 (en) 2006-01-25 2007-11-15 Insys Therapeutics, Inc. Sublingual fentanyl spray
US20090124554A1 (en) 1997-10-01 2009-05-14 Dugger Iii Harry A Buccal, polar and non-polar spray or capsule containing drugs for treating pain
US20090162300A1 (en) 1997-10-01 2009-06-25 Dugger Iii Harry A Buccal, polar and non-polar spray containing alprazolam

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0305579D0 (en) 2003-03-11 2003-04-16 Dallas Burston Ltd Pharmaceutical compositions
US8535714B2 (en) * 2006-01-06 2013-09-17 Acelrx Pharmaceuticals, Inc. Small volume oral transmucosal dosage forms containing sufentanil for treatment of pain

Patent Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4244478A (en) 1979-06-27 1981-01-13 Mpl, Inc. Closure assembly for unit dose vial
WO1990007333A1 (en) 1989-01-06 1990-07-12 Riker Laboratories, Inc. Fentanyl containing aerosol compositions
US5219083A (en) 1992-04-02 1993-06-15 Sterling Winthrop Inc. Stopper for reduction of particulate matter
RU2156126C2 (en) 1993-08-10 2000-09-20 Дзе Бутс Компани ПЛС Therapeutically ready medicinal form
US5976504A (en) 1993-08-10 1999-11-02 The Boots Company, Plc Topical aerosol formulation for cooling of mammalian tissues
US5958379A (en) 1994-09-30 1999-09-28 Mika Pharma Gesellschaft Fuer Die Entwicklung Und Vermarktung Pharmazeutischer Producte Mbh Pharmaceutical composition
US20040265239A1 (en) 1997-10-01 2004-12-30 Novadel Pharma Inc. Buccal, polar and non-polar spray containing zolpidem
US20040136915A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing atropine
US20090162300A1 (en) 1997-10-01 2009-06-25 Dugger Iii Harry A Buccal, polar and non-polar spray containing alprazolam
US20090124554A1 (en) 1997-10-01 2009-05-14 Dugger Iii Harry A Buccal, polar and non-polar spray or capsule containing drugs for treating pain
US20030039680A1 (en) 1997-10-01 2003-02-27 Flemington Pharmaceutical Corporation Buccal, polar and non-polar spray or capsule
US20030077228A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating endocrine disorders
US20030077229A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing cardiovascular or renal drugs
US20030082107A1 (en) 1997-10-01 2003-05-01 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating an infectious disease or cancer
US20030095926A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the gastrointestinal tract or urinary tract
US20030095925A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating metabolic disorders
US20030095927A1 (en) 1997-10-01 2003-05-22 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating muscular and skeletal disorders
US20050287075A1 (en) 1997-10-01 2005-12-29 Dugger Harry A Iii Buccal, polar and non-polar spray or capsule containing drugs for treating pain
US20030190286A1 (en) 1997-10-01 2003-10-09 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating allergies or asthma
US20050281753A1 (en) 1997-10-01 2005-12-22 Dugger Harry A Iii Buccal, polar and non-polar spray or capsule
US20050281752A1 (en) 1997-10-01 2005-12-22 Dugger Harry A Iii Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the central nervous system
US20040120895A1 (en) 1997-10-01 2004-06-24 Novadel Pharma, Inc. Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the central nervous system
US20050180923A1 (en) 1997-10-01 2005-08-18 Dugger Harry A.Iii Buccal, polar and non-polar spray containing testosterone
US20040136913A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing sumatriptan
US20040136914A1 (en) 1997-10-01 2004-07-15 Dugger Harry A. Buccal, polar and non-polar spray containing ondansetron
US20050163719A1 (en) 1997-10-01 2005-07-28 Dugger Harry A.Iii Buccal, polar and non-polar spray containing diazepam
US20050002867A1 (en) 1997-10-01 2005-01-06 Novadel Pharma Inc. Buccal, polar and non-polar sprays containing propofol
US20040141923A1 (en) 1997-10-01 2004-07-22 Dugger Harry A. Buccal, polar and non-polar spray containing alprazolam
US6759059B1 (en) 1998-09-24 2004-07-06 Diabact Ab Fentanyl composition for the treatment of acute pain
RU2232580C2 (en) 1998-09-24 2004-07-20 Диабакт Аб Fentanyl composition for treatment of acute pain
US20020055496A1 (en) 1999-02-12 2002-05-09 Mccoy Randall Formulation and system for intra-oral delivery of pharmaceutical agents
WO2000047203A1 (en) 1999-02-12 2000-08-17 Mqs, Inc. Formulation and system for intra-oral delivery of pharmaceutical agents
US20030190290A1 (en) 2000-06-22 2003-10-09 Calvin Ross Pharmaceutical compositions
WO2001097780A2 (en) 2000-06-22 2001-12-27 Pharmasol Ltd Pharmaceutical compositions comprising an opioid analgesic
US20020160991A1 (en) 2000-12-04 2002-10-31 Liming Shao Orally-bioavailable formulations of fentanyl and congeners thereof
US20040092428A1 (en) 2001-11-27 2004-05-13 Hongming Chen Oral pharmaceuticals formulation comprising paclitaxel, derivatives and methods of administration thereof
US6946150B2 (en) * 2002-08-14 2005-09-20 Gw Pharma Limited Pharmaceutical formulation
WO2004016243A2 (en) 2002-08-16 2004-02-26 Generex Pharmaceuticals Inc. Methods of administering and enhancing absorption of pharmaceutical agents
WO2004075877A1 (en) 2003-02-24 2004-09-10 Pharmaceutical Productions, Inc. Transmucosal drug delivery system
US20060062812A1 (en) 2003-03-11 2006-03-23 Calvin Ross Novel compositions
WO2004080382A2 (en) 2003-03-11 2004-09-23 Arakis Ltd. Novel compositions containing fentanyl
WO2007007059A1 (en) 2005-07-08 2007-01-18 Sosei R & D Ltd. Fentanyl formulation containing an essential oil
US20070261695A1 (en) 2006-01-25 2007-11-15 Insys Therapeutics, Inc. Sublingual fentanyl spray

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
An English translation of the Russian Examination Report dated Aug. 2009 from corresponding Russian Application No. 2008130763.
Examination Report dated Aug. 6, 2009 from corresponding Australian Application No. 2007208229.
Examination Report dated Mar. 18, 2010 from corresponding New Zealand Application No. 569949.
Examination Report dated May 5, 2010 from corresponding Canadian Application No. 2,637,672.
Examination Report for corresponding Australian Patent Application No. 20008282743, mailed on Feb. 9, 2011.
Examination Report for corresponding New Zealand Application No. 583651, mailed on Nov. 15, 2010.
International Preliminary Report on Patentability dated Sep. 4, 2008 from corresponding Int'l Application No. PCT/US07/02163.
Int'l Preliminary Report on Patentability from related Int'l Application No. PCT/US08/09359 dated Feb. 2, 2010.
ISR and Written Opinion from related Int'l Application No. PCT/US08/09359 dated Jan. 9, 2009.
Lejus, et al., "Fentanyl versus sufentanil: plasma concentrations during continuous epidural postoperative infusion in children," British Journal of Anaesthesia, vol. 85, Issue 4, Oct. 2000, pp. 615-617.
Marier, J-F., et al. "Comparative bioequivalence study between a novel matrix transdermal delivery system of fentanyl and a commercially available reservoir formulation", Br. J. Clin. Pharmacol. Aug. 2006, vol. 63, No. 1, pp. 121-124, esp. p. 123, Figure 1.
Mathar, L.E., et al. "Pulmonary administration of aerosolised fentanyl: pharmacokinetic analysis of systemic delivery" Br. J. Clin. Pharmacol. Jan. 1998, vol. 46, pp. 37-43.
Office Action on Patent Application No. 200780003555.X dated Jul. 9, 2010.
Smyth, et al, 2003, AAPS PharmSciTech 2003; 4 (3): 1-11.
Supplementary European Search Report, 2 pages, for corresponding European Patent Application No. 07762549.9, mailed on Sep. 30, 2010.
The International Search Report and Written Opinion dated Jul. 11, 2008 from corresponding Int'l Application No. PCT/US07/02163.
U.S. Appl. No. 11/698,739 Office Action Summary dated Sep. 15, 2010.

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8835460B2 (en) 2006-01-25 2014-09-16 Insys Therapeutics, Inc. Sublingual fentanyl spray and methods of treating pain
US9289387B2 (en) 2006-01-25 2016-03-22 Insys Development Company, Inc. Method of treating pain by administering sublingual fentanyl spray
US9642797B2 (en) 2006-01-25 2017-05-09 Insys Development Company, Inc. Sublingual fentanyl spray and methods of use to treat pain
US10016403B2 (en) 2006-01-25 2018-07-10 Insys Development Company, Inc. Sublingual fentanyl spray
US10265309B2 (en) * 2016-05-27 2019-04-23 Insys Development Company, Inc. Sublingual opioid formulations containing naloxone
US10874658B2 (en) * 2016-05-27 2020-12-29 Hikma Pharmaceuticals Usa Inc. Sublingual opioid formulations containing naloxone

Also Published As

Publication number Publication date
US10610523B2 (en) 2020-04-07
EP2180844A4 (en) 2013-09-11
US20170239230A1 (en) 2017-08-24
US20140343288A1 (en) 2014-11-20
DK2180844T3 (en) 2018-04-23
US20160095846A1 (en) 2016-04-07
US20130296368A1 (en) 2013-11-07
WO2009017837A9 (en) 2009-06-25
CA2698749A1 (en) 2009-02-05
PL2180844T3 (en) 2018-07-31
US9241935B2 (en) 2016-01-26
AU2008282743B2 (en) 2012-11-29
EP2180844B1 (en) 2018-02-21
US9642844B2 (en) 2017-05-09
ES2668366T3 (en) 2018-05-17
EP2180844A2 (en) 2010-05-05
WO2009017837A3 (en) 2009-04-30
NO2180844T3 (en) 2018-07-21
CA2698749C (en) 2017-05-23
AU2008282743A1 (en) 2009-02-05
PT2180844T (en) 2018-04-06
WO2009017837A2 (en) 2009-02-05
US20090176834A1 (en) 2009-07-09
US8835459B2 (en) 2014-09-16

Similar Documents

Publication Publication Date Title
US10610523B2 (en) Sublingual fentanyl spray
US10016403B2 (en) Sublingual fentanyl spray
DK2643092T3 (en) Cyclic product supply by a centrifuge and centrifuge thereto

Legal Events

Date Code Title Description
AS Assignment

Owner name: INSYS THERAPEUTICS, INC., ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KOTTAYIL, S. GEORGE;GOSKONDA, VENKAT R.;ZHU, ZHONGYUAN;AND OTHERS;REEL/FRAME:022475/0834;SIGNING DATES FROM 20090305 TO 20090328

Owner name: INSYS THERAPEUTICS, INC., ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KOTTAYIL, S. GEORGE;GOSKONDA, VENKAT R.;ZHU, ZHONGYUAN;AND OTHERS;SIGNING DATES FROM 20090305 TO 20090328;REEL/FRAME:022475/0834

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: INSYS PHARMA, INC., ARIZONA

Free format text: CHANGE OF NAME;ASSIGNOR:INSYS THERAPEUTICS, INC.;REEL/FRAME:033596/0258

Effective date: 20110120

AS Assignment

Owner name: INSYS DEVELOPMENT COMPANY, INC., ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INSYS PHARMA, INC.;REEL/FRAME:037652/0049

Effective date: 20160202

FEPP Fee payment procedure

Free format text: PAT HOLDER NO LONGER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: STOL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: BTCP PHARMA, LLC, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:INSYS THERAPEUTICS, INC.;INSYS DEVELOPMENT COMPANY, INC.;REEL/FRAME:050657/0793

Effective date: 20190926

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8